Sodium nitrate alleviates functional muscle ischaemia in patients with Becker muscular dystrophy

Michael D Nelson, Ryan Rosenberry, Rita Barresi, Evgeny I Tsimerinov, Florian Rader, Xiu Tang, O'Neil Mason, Avery Schwartz, Thomas Stabler, Sarah Shidban, Neigena Mobaligh, Shomari Hogan, Robert Elashoff, Jason D Allen, Ronald G Victor, Michael D Nelson, Ryan Rosenberry, Rita Barresi, Evgeny I Tsimerinov, Florian Rader, Xiu Tang, O'Neil Mason, Avery Schwartz, Thomas Stabler, Sarah Shidban, Neigena Mobaligh, Shomari Hogan, Robert Elashoff, Jason D Allen, Ronald G Victor

Abstract

Becker muscular dystrophy (BMD) is a progressive X-linked muscle wasting disease for which there is no treatment. BMD is caused by in-frame mutations in the gene encoding dystrophin, a structural cytoskeletal protein that also targets other proteins to the sarcolemma. Among these is neuronal nitric oxide synthase mu (nNOSμ), which requires specific spectrin-like repeats (SR16/17) in dystrophin's rod domain and the adaptor protein α-syntrophin for sarcolemmal targeting. When healthy skeletal muscle is exercised, sarcolemmal nNOSμ-derived nitric oxide (NO) attenuates α-adrenergic vasoconstriction, thus optimizing perfusion. In the mdx mouse model of dystrophinopathy, this protective mechanism (functional sympatholysis) is defective, resulting in functional muscle ischaemia. Treatment with a NO-donating non-steroidal anti-inflammatory drug (NSAID) alleviates this ischaemia and improves the murine dystrophic phenotype. In the present study, we report that, in 13 men with BMD, sympatholysis is defective mainly in patients whose mutations disrupt sarcolemmal targeting of nNOSμ, with the vasoconstrictor response measured as a decrease in muscle oxygenation (near infrared spectroscopy) to reflex sympathetic activation. Then, in a single-arm, open-label trial in 11 BMD patients and a double-blind, placebo-controlled cross-over trial in six patients, we show that acute treatment with oral sodium nitrate, an inorganic NO donor without a NSIAD moiety, restores sympatholysis and improves post-exercise hyperaemia (Doppler ultrasound). By contrast, sodium nitrate improves neither sympatholysis, nor hyperaemia in healthy controls. Thus, a simple NO donor recapitulates the vasoregulatory actions of sarcolemmal nNOS in BMD patients, and constitutes a putative novel therapy for this disease.

© 2015 The Authors. The Journal of Physiology © 2015 The Physiological Society.

Figures

Figure 1. BMD mutations that interrupt s…
Figure 1. BMD mutations that interrupt sarcolemmal anchoring of nNOS impair functional sympatholysis
Left: representative immunohistochemistry from muscle biopsies from a healthy control subject (A) and two BMD patients (B and C). Muscle biopsy sections stained with haematoxylin and eosin (upper left) from the two BMD patients show typical dystrophic hallmarks, including variation in muscle fibre size, internal nuclei, fibrosis and an increase in fat tissue. Staining for dystrophin C‐terminus (DYS‐C) is present in both BMD patients, although no staining for dystrophin N‐terminus (DYS‐N) is present (evidence for truncated dystrophin). Sarcolemmal nNOS staining is absent in (B) but present in (C). Middle: representative tracing from a healthy control subject (A), a BMD patient lacking sarcolemmal nNOS (B, P2) and a BMD patient with nNOS retained at the sarcolemma (C, P11). A black circle highlights the LBNP‐induced change in forearm muscle oxygenation at rest, whereas a red circle highlights the LBNP‐induced change in forearm muscle oxygenation during exercise. Note that, in healthy controls, the decrease in forearm muscle oxygenation is greatly attenuated (termed functional sympatholysis). At the end of each experiment, an arm cuff was inflated to suprasystolic pressure to occlude the forearm circulation, producing a maximal decrease in forearm muscle oxygenation to calculate the TLS. Right: summary forearm muscle oxygenation data from 13 healthy subjects (A), seven BMD patients in whom sarcolemmal nNOS was absent (B) and six BMD patients in whom sarcolemmal nNOS was present (C). Data are reported as the mean + SE.
Figure 2. Sodium nitrate alleviates functional muscle…
Figure 2. Sodium nitrate alleviates functional muscle ischaemia in BMD: acute treatment trial
A, representative tracing (left) from a patient with BMD, showing that sympatholysis is impaired because handgrip fails to attenuate the LBNP‐induced reflex decrease in forearm muscle oxygenation (black circle). At the end of each experiment, an arm cuff was inflated to suprasystolic pressure to occlude the forearm circulation, producing a maximal decrease in forearm muscle oxygenation to calculate the TLS. Right: summary forearm muscle oxygenation data from the 10 patients with BMD studied at rest (black bar) and during exercise (grey bar), expressed as a percentage of TLS. B, representative tracing from the same BMD subject after oral treatment with sodium nitrate, showing that the LBNP‐induced decrease in forearm muscle oxygenation is greatly attenuated during mild handgrip exercise (grey circle). Summary data are shown on the right (n = 10). Of note, we observed no difference between the LBNP response at rest before and after sodium nitrate (not statistically significant). Data are reported as the mean + SE.
Figure 3. Patient‐specific data from the acute…
Figure 3. Patient‐specific data from the acute treatment trial
A, at baseline, the LBNP‐induced decrease in forearm muscle oxygenation (HbO2 + MbO2) is very consistent at rest (black bar) and during exercise (grey bar) for each patient. B, sodium nitrate restored functional sympatholysis in all but two subjects (P1 and P13). Note that Patient 5 (P5) deviated from the protocol by brushing his teeth during this initial study visit (V1). Accordingly, his data were removed from the analysis. When we repeated the experiment in P11 several months later (V2) (i.e. when the subject was reminded to avoid using toothpaste and mouthwash), the change in plasma nitrite concentration doubled from the first visit, and sympatholysis was improved. Data are expressed as a percentage of the TLS.
Figure 4. Test–retest reproducibility
Figure 4. Test–retest reproducibility
Four BMD subjects were studied several weeks apart. The data show excellent test–retest reproducibility for functional sympatholysis before sodium nitrate ingestion (showing that functional muscle ischaemia is consistently impaired in four of four patients) and after sodium nitrate ingestion (showing consistent restoration of functional sympatholysis in four of four patients).
Figure 5. Placebo‐controlled cross‐over trial: sodium nitrate…
Figure 5. Placebo‐controlled cross‐over trial: sodium nitrate alleviates functional muscle ischaemia and increased exercise‐induced hyperaemia in BMD
A, sodium nitrate supplementation restored functional sympatholysis (exercise‐induced attenuation of reflex vasoconstriction, measured by NIR) in men with BMD (n = 6), whereas placebo had no such effect. B, sodium nitrate supplementation does not affect resting skeletal muscle blood flow (measured by brachial artery Doppler ultrasound) but significantly increases exercise‐induced hyperaemia compared to placebo (n = 6). Data are expressed as the mean ± SE.
Figure 6. Sodium nitrate does not improve…
Figure 6. Sodium nitrate does not improve functional sympatholysis and does not increase exercise‐induced hyperaemia in healthy volunteers
A, sodium nitrate supplementation failed to improve functional sympatholysis (exercise‐induced attenuation of reflex vasoconstriction, measured by NIR) in healthy male volunteers (n = 8). B, sodium nitrate supplementation did not affect resting skeletal muscle blood flow (measured by brachial artery Doppler ultrasound), nor did it affect exercise‐induced hyperaemia (n = 8). Data are expressed as the mean ± SE.
Figure 7. Circulating nitrite serves as an…
Figure 7. Circulating nitrite serves as an alternative nitric oxide donor only in the presence of deoxyHb and/or deoxyMb
A, rhythmic handgrip exercise significantly increases skeletal muscle deoxyHb and deoxyMb content, as measured by NIR over the flexor digitorum profundus muscle (n = 6). Insert: original NIR record, showing a steep increase in deoxyHb content during the transition from rest to exercise. All data are expressed as a percentage of the TLS. B, sodium nitrate significantly increases circulating plasma nitrite concentration in patients with BMD (n = 6). C, circulating NO2 −is reduced to NO via deoxyHb in the microvascular space, and by deoxyMb in skeletal myocytes, leading to improvements in skeletal muscle blood flow regulation. Data are reported as the mean + SE.

References

    1. Adamo CM, Dai DF, Percival JM, Minami E, Willis MS, Patrucco E, Froehner SC & Beavo JA (2010). Sildenafil reverses cardiac dysfunction in the mdx mouse model of Duchenne muscular dystrophy. Proc Natl Acad Sci USA 107, 19079–19083.
    1. Anderson JE (2000). A role for nitric oxide in muscle repair: nitric oxide‐mediated activation of muscle satellite cells. Mol Biol Cell 11, 1859–1874.
    1. Anderson LVB & Davison K (1999). Multiplex western blotting system for the analysis of muscular dystrophy proteins. Am J Pathol 154, 1017–1022.
    1. Asai A, Sahani N, Kaneki M, Ouchi Y, Martyn JAJ & Yasuhara SE (2007). Primary role of functional ischemia, quantitative evidence for the two‐hit mechanism, and phosphodiesterase‐5 inhibitor therapy in mouse muscular dystrophy. PLoS ONE 2, e806.
    1. Brenman JE, Chao DS, Xia H, Aldape K & Bredt DS (1995). Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy. Cell 82, 743–752.
    1. Brunelli S, Sciorati C, D'Antona G, Innocenzi A, Covarello D, Galvez BG, Perrotta C, Monopoli A, Sanvito F, Bottinelli R, Ongini E, Cossu G & Clementi E (2007). Nitric oxide release combined with nonsteroidal antiinflammatory activity prevents muscular dystrophy pathology and enhances stem cell therapy. Proc Natl Acad Sci USA 104, 264–269.
    1. Buono R, Vantaggiato C, Pisa V, Azzoni E, Bassi MT, Brunelli S, Sciorati C & Clementi E (2012). Nitric oxide sustains long‐term skeletal muscle regeneration by regulating fate of satellite cells via signaling pathways requiring Vangl2 and cyclic GMP. Stem Cells 30, 197–209.
    1. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, Kaul A, Kinnett K, McDonald C, Pandya S, Poysky J, Shapiro F, Tomezsko J & Constantin C (2010. a). Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management. Lancet Neurol 9, 77–93.
    1. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, Kaul A, Kinnett K, McDonald C, Pandya S, Poysky J, Shapiro F, Tomezsko J & Constantin C (2010. b). Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care. Lancet Neurol 9, 177–189.
    1. Chao DS, Gorospe JR, Brenman JE, Rafael JA, Peters MF, Froehner SC, Hoffman EP, Chamberlain JS & Bredt DS (1996). Selective loss of sarcolemmal nitric oxide synthase in Becker muscular dystrophy.J Exp Med 184, 609–618.
    1. Chavoshan B, Sander M, Sybert TE, Hansen J, Victor RG & Thomas GD (2002). Nitric oxide‐dependent modulation of sympathetic neural control of oxygenation in exercising human skeletal muscle. J Physiol 540, 377–386.
    1. Clifford PS & Hellsten Y (2004). Vasodilatory mechanisms in contracting skeletal muscle. J Appl Physiol 97, 393–403.
    1. Cordani N, Pisa V, Pozzi L, Sciorati C & Clementi E (2014). Nitric oxide controls fat deposition in dystrophic skeletal muscle by regulating fibro‐adipogenic precursor differentiation. Stem Cells 32, 874–885.
    1. Dinenno FA & Joyner MJ (2003). Blunted sympathetic vasoconstriction in contracting skeletal muscle of healthy humans: is nitric oxide obligatory? J Physiol 553, 281–292.
    1. Duan D (2006). Challenges and opportunities in dystrophin‐deficient cardiomyopathy gene therapy. Hum Mol Genet 15, R253–R261.
    1. Eckel RH, Jakicic JM, Ard JD, de Jesus JM, Houston Miller N, Hubbard VS, Lee IM, Lichtenstein AH, Loria CM, Millen BE, Nonas CA, Sacks FM, Smith J, Svetkey LP, Wadden TA & Yanovski SZ (2014). 2013 AHA/ACC guideline on lifestyle management to reduce cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force onPractice Guidelines. J Am Coll Cardiol 63, 2960–2984.
    1. Emery AE (2002). The muscular dystrophies. Lancet 359, 687–695.
    1. Fadel PJ, Farias M III, Gallagher KM, Wang Z & Thomas GD (2012). Oxidative stress and enhanced sympathetic vasoconstriction in contracting muscles of nitrate‐tolerant rats and humans. J Physiol 590, 395–407.
    1. Flanigan KM, Niederhausern AV, Dunn DM, Alder J, Mendell JR & Weiss RB (2003). Rapid direct sequence analysis of the dystrophin gene. Am J Hum Genet 72, 931–939.
    1. Gentil C, Leturcq F, Ben Yaou R, Kaplan JC, Laforet P, Penisson‐Besnier I, Espil‐Taris C, Voit T, Garcia L & Pietri‐Rouxel F (2012). Variable phenotype of del45‐55 Becker patients correlated with nNOSμ mislocalization and RYR1 hypernitrosylation. Hum Mol Genet 21, 3449–3460.
    1. Govoni M, Jansson E, Weitzberg E & Lundberg JO (2008). The increase in plasma nitrite after a dietary nitrate load is markedly attenuated by an antibacterial mouthwash. Nitric Oxide 19, 333–337.
    1. Gratton E, Fantini S, Franceschini MA, Gratton G & Fabiani M (1997). Measurements of scattering and absorption changes in muscle and brain. Philos Trans R Soc Lond B Biol Sci 352, 727–735.
    1. Hansen J, Thomas GD, Harris SA, Parsons WJ & Victor RG (1996). Differential sympathetic neural control of oxygenation in resting and exercising human skeletal muscle. J Clin Invest 98, 584–596.
    1. Hendgen‐Cotta UB, Luedike P, Totzeck M, Kropp M, Schicho A, Stock P, Rammos C, Niessen M, Heiss C, Lundberg JO, Weitzberg E, Kelm M & Rassaf T (2012). Dietary nitrate supplementation improves revascularization in chronic ischemia. Circulation 126, 1983–1992.
    1. Herr MD, Hogeman CS, Koch DW, Krishnan A, Momen A & Leuenberger UA (2010). A real‐time device for converting Doppler ultrasound audio signals into fluid flow velocity. Am J Physiol Heart Circ Physiol 298, H1626–H1632.
    1. Jobsis FF (1977). Noninvasive, infrared monitoring of cerebral and myocardial oxygen sufficiency and circulatory parameters. Science 198, 1264–1267.
    1. Kameya S, Miyagoe Y, Nonaka I, Ikemoto T, Endo M, Hanaoka K, Nabeshima YI & Takeda S (1999). Alpha 1‐syntrophin gene disruption results in the absence of neuronal‐type nitric‐oxide synthase at the sarcolemma but does not induce muscle degeneration. J Biol Chem 274, 2193–2200.
    1. Kenjale AA, Ham KL, Stabler T, Robbins JL, Johnson JL, VanBruggen M, Privette G, Yim E, Kraus WE & Allen JD (2011). Dietary nitrate supplementation enhances exercise performance in peripheral arterial disease. J Appl Physiol 110, 1582–1591.
    1. Khairallah M, Khairallah RJ, Young ME, Allen BG, Gillis MA, Danialou G, Deschepper CF, Petrof BJ & Des Rosiers C (2008). Sildenafil and cardiomyocyte‐specific cGMP signaling prevent cardiomyopathic changes associated with dystrophin deficiency. Proc Natl Acad Sci USA 105, 7028–7033.
    1. Kobayashi YM, Rader EP, Crawford RW, Iyengar NK, Thedens DR, Faulkner JA, Parikh SV, Weiss RM, Chamberlain JS, Moore SA & Campbell KP (2008). Sarcolemma‐localized nNOS is required to maintain activity after mild exercise. Nature 456, 511–515.
    1. Lai Y, Thomas GD, Yue Y, Yang HT, Li D, Long C, Judge L, Bostick B, Chamberlain JS, Terjung RL & Duan D (2009). Dystrophins carrying spectrin‐like repeats 16 and 17 anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of muscular dystrophy. J Clin Invest 119, 624–635.
    1. Larsen FJ, Ekblom BR, Sahlin K, Lundberg JO & Weitzberg E (2006). Effects of dietary nitrate on blood pressure in healthy volunteers. N Engl J Med 355, 2792–2793.
    1. Li D, Yue Y, Lai Y, Hakim CH & Duan D (2011). Nitrosative stress elicited by nNOS delocalization inhibits muscle force in dystrophin‐null mice. J Pathol 223, 88–98.
    1. Lundberg JO, Weitzberg E & Gladwin MT (2008). The nitrate‐nitrite‐nitric oxide pathway in physiology and therapeutics. Nat Rev Drug Discov 7, 156–167.
    1. Marques MJ, Luz MAM, Minatel E & Neto HS (2005). Muscle regeneration in dystrophic mdx mice is enhanced by isosorbide dinitrate. Neurosci Lett 382, 342–345.
    1. Martin EA, Barresi R, Byrne BJ, Tsimerinov EI, Scott BL, Walker AE, Gurudevan SV, Anene F, Elashoff RM, Thomas GD & Victor RG (2012). Tadalafilalleviates muscle ischemia in patients with becker muscular dystrophy. Sci Trans Med 4, 162ra155.
    1. Monaco AP (1989). Dystrophin, the protein product of the Duchenne/Becker muscular dystrophy gene. Trends Biochem Sci 14, 412–415.
    1. Nelson MD, Rader F, Tang X, Tavyev J, Nelson SF, Miceli MC, Elashoff RM, Sweeney HL & Victor RG (2014). PDE5 inhibition alleviates functional muscle ischemia in boys with Duchenne muscular dystrophy. Neurology 82, 2085–2091.
    1. Percival JM, Whitehead NP, Adams ME, Adamo CM, Beavo JA & Froehner SC (2012). Sildenafil reduces respiratory muscle weakness and fibrosis in the mdx mouse model of Duchenne muscular dystrophy. J Pathol 228, 77–87.
    1. Price A, Raheja P, Wang Z, Arbique D, Adams‐Huet B, Mitchell JH, Victor RG, Thomas GD & Vongpatanasin W (2013). Differential effects of nebivolol versus metoprolol on functional sympatholysis in hypertensive humans. Hypertension 61, 1263–1269.
    1. Prosser BL, Khairallah RJ, Ziman AP, Ward CW & Lederer WJ (2013). X‐ROS signaling in the heart and skeletal muscle: stretch‐dependent local ROS regulates [Ca2+]i. J Mol Cell Cardiol 58, 172–181.
    1. Sander M, Chavoshan B, Harris SA, Iannaccone ST, Stull JT, Thomas GD & Victor RG (2000). Functional muscle ischemia in neuronal nitric oxide synthase‐deficient skeletal muscle of children with Duchenne muscular dystrophy.Proc Natl Acad Sci USA 97, 13818–13823.
    1. Sciorati C, Miglietta D, Buono R, Pisa V, Cattaneo D, Azzoni E, Brunelli S & Clementi E (2011). A dual acting compound releasing nitric oxide (NO) and ibuprofen, NCX 320, shows significant therapeutic effects in a mouse model of muscular dystrophy. Pharmacol Res 64, 210–217.
    1. Shiva S, Huang Z, Grubina R, Sun J, Ringwood LA, MacArthur PH, Xu X, Murphy E, Darley‐Usmar VM & Gladwin MT (2007). Deoxymyoglobinis a nitrite reductase that generates nitric oxide and regulates mitochondrial respiration. Circ Res 100, 654–661.
    1. Siervo M, Lara J, Ogbonmwan I & Mathers JC (2013). Inorganic nitrate and beetroot juice supplementation reduces blood pressure in adults: a systematic review and meta‐analysis. J Nutr 143, 818–826.
    1. Thomas GD, Hansen J & Victor RG (1994). Inhibition of alpha 2‐adrenergic vasoconstriction during contraction of glycolytic, not oxidative, rat hindlimb muscle. Am J Physiol Heart Circ Physiol 266, H920–H929.
    1. Thomas GD, Sander M, Lau KS, Huang PL, Stull JT & Victor RG (1998). Impaired metabolic modulation of alpha‐adrenergic vasoconstriction in dystrophin‐deficient skeletal muscle.Proc Natl Acad Sci USA 95, 15090–15095.
    1. Thomas GD, Shaul PW, Yuhanna IS, Froehner SC & Adams ME (2003). Vasomodulation by skeletal muscle‐derived nitric oxide requires alpha‐syntrophin‐mediated sarcolemmal localization of neuronal nitric oxide synthase. Circ Res 92, 554–560.
    1. Thomas GD & Victor RG (1998). Nitric oxide mediates contraction‐induced attenuation of sympathetic vasoconstriction in rat skeletal muscle. J Physiol 506, 817–826.
    1. Thomas GD, Ye J, De Nardi C, Monopoli A, Ongini E & Victor RG (2012). Treatment with a nitric oxide‐donating NSAID alleviates functional muscle ischemia in the mouse model of Duchenne muscular dystrophy. PLoS ONE 7, e49350.
    1. Thomas GD, Zhang W & Victor RG (2001). Impaired modulation of sympathetic vasoconstriction in contracting skeletal muscle of rats with chronic myocardial infarctions: role of oxidative stress. Circ Res 88, 816–823.
    1. Tidball JG & Wehling‐Henricks M (2004). Expression of a NOS transgene in dystrophin‐deficient muscle reduces muscle membrane damage without increasing the expression of membrane‐associated cytoskeletal proteins. Mol Genet Metab 82, 312–320.
    1. Torelli S, Brown S, Jimenez‐Mallebrera C, Feng L, Muntoni F & Sewry C (2004). Absence of neuronal nitric oxide synthase (nNOS) as a pathological marker for the diagnosis of Becker muscular dystrophy with rod domain deletions. Neuropathol Appl Neurobiol 30, 540–545.
    1. Uaesoontrachoon K, Quinn JL, Tatem KS, Van Der Meulen JH, Yu Q, Phadke A, Miller BK, Gordish‐Dressman H, Ongini E, Miglietta D & Nagaraju K (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.
    1. Vongpatanasin W, Wang Z, Arbique D, Arbique G, Adams‐Huet B, Mitchell JH, Victor RG & Thomas GD (2011). Functional sympatholysis is impaired in hypertensive humans. J Physiol 589, 1209–1220.
    1. Wylie LJ, Kelly J, Bailey SJ, Blackwell JR, Skiba PF, Winyard PG, Jeukendrup AE, Vanhatalo A & Jones AM (2013). Beetroot juice and exercise: pharmacodynamic and dose–response relationships. J Appl Physiol 115, 325–336.
    1. Zhao W, Swanson SA, Ye J, Li X, Shelton JM, Zhang W & Thomas GD (2006). Reactive oxygen species impair sympathetic vasoregulation in skeletal muscle in angiotensin II‐dependent hypertension. Hypertension 48, 637–643.

Source: PubMed

3
Subskrybuj