• No results found

VU Research Portal

N/A
N/A
Protected

Academic year: 2021

Share "VU Research Portal"

Copied!
25
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Mechanisms leading to muscle weakness in Nemaline myopathy Joureau, B.

2019

document version

Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA)

Joureau, B. (2019). Mechanisms leading to muscle weakness in Nemaline myopathy.

General rights

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights. • Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain

• You may freely distribute the URL identifying the publication in the public portal ?

Take down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address:

(2)

CHAPTER

Muscle weakness in respiratory

and peripheral skeletal muscles in

a mouse model for

nebulin-based nemaline myopathy

Barbara Joureau, J.M. de Winter, K. Stam, H. Granzier, and C.A.C. Ottenheijm

Published as Research Article Neuromuscul Disord. 2017 Jan;27(1):83-89

(3)
(4)

ABSTRACT

Nemaline myopathy is among the most common non-dystrophic congenital myopathies, and is characterized by the presence of nemaline rods in skeletal muscles fibers, general muscle weakness, and hypotonia. Although respiratory failure is the main cause of death in nemaline myopathy, only little is known regarding the contractile strength of the diaphragm, the main muscle of inspiration. To investigate diaphragm contractility, in the present study we took advantage of a mouse model for nebulin-based nemaline myopathy that we recently developed. In this mouse model, exon 55 of Neb is deleted (NebΔExon55), a mutation frequently

found in patients. Diaphragm contractility was determined in permeabilized muscle fibers and was compared to the contractility of permeabilized fibers from three peripheral skeletal muscles: soleus, extensor digitorum longus, and gastrocnemius. The force generating capacity of diaphragm muscle fibers of NebΔExon55 mice

was reduced to 25% of wildtype levels, indicating severe contractile weakness. The contractile weakness of diaphragm fibers was more pronounced than that observed in soleus muscle, but not more pronounced than that observed in extensor digitorum longus and gastrocnemius muscles. The reduced muscle contractility was at least partly caused by changes in cross-bridge cycling kinetics which reduced the number of bound cross-bridges. The severe diaphragm weakness likely contributes to the development of respiratory failure in NebΔExon55 mice and might explain their early,

postnatal death.

(5)
(6)

INTRODUCTION

Nemaline myopathy (NM) is a non-dystrophic congenital myopathy described for the first time in the 1960s1. Eleven genes have been implicated in NM. Six of these genes code for components of the skeletal muscle thin filament: alpha actin 1 (ACTA1)2, alpha- and beta-tropomyosin (TPM3 and TPM2)3, 4, nebulin (NEB)5, leiomodin-3 (LMOD3)6 and troponin T (TNNT1)7. Four genes code for proteins which play a role in the regulation and the stability of the thin filament: cofilin 2 (CFL2)8 regulate actin filament dynamics; kelch family member 40 (KLHL40) is implicated in myogenesis9 and in the stabilization of thin filament proteins9, 10 and kelch family member 41 (KLHL41) modulates ubiquitination of thin filament proteins10, 11. The role of Kelch repeat and BTB (POZ) Domain Containing 13 (KBTBD13) is unclear, but might involve a role in the ubiquitin-proteasome pathway12. The most recent implicated gene, MYO18B, codes for an unconventional myosin heavy chain expressed in cardiac and skeletal muscles. Its function is incompletely understood13.

NM affects skeletal muscles, including the muscles of respiration. Consequently, patients with NM have reduced spirometric values such as forced vital capacity, peak expiratory flow, forced expiratory volume in 1 second14-16, and they may suffer from the sensation of dyspnea15-19. Respiratory failure is the main cause of death in NM20, and occurs even in ambulant patients who otherwise appear to be only mildly affected; respiratory failure may even be the presenting feature. This suggests that weakness of respiratory muscles may be much greater than that of peripheral skeletal muscles. The pathophysiology of respiratory muscle weakness is not completely understood, but might involve a dysfunctional diaphragm. The diaphragm is the main muscle of inspiration, and weakness of the diaphragm impairs respiration and causes dyspnea21. Thus, insights in diaphragm contractility in NM is important for developing treatment strategies.

The little information available regarding diaphragm contractility

(7)

in NM is a consequence of technical and ethical limitations in assessing diaphragm contractility in vivo and in obtaining diaphragm muscle biopsies from patients. Therefore, in the present study we took advantage of a mouse model for nebulin-based NM that we recently developed22. Mutations in NEB account for more than 50% of the gene mutations reported in NM patients23, and in this mouse model exon 55 of Neb is deleted a mutation frequently found in patients22. To assess diaphragm contractility, we isolated permeabilized muscle fibers from the diaphragm and activated the fibers with calcium. Muscle fiber contractility was determined and compared to the contractility of muscle fibers of three peripheral skeletal muscles: soleus, extensor digitorum longus and gastrocnemius. Our results indicate that diaphragm muscle fibers of NebΔExon55 mice exhibit severe contractile weakness, a

weakness that was more pronounced than that observed in soleus muscle, but not more pronounced than that in extensor digitorum longus (EDL) and gastrocnemius muscles.

MATERIAL & METHODS

Generation of NebΔExon55 mice

The procedure for the generation of the NebΔExon55 mice has been

previously described22.

Muscle preparations

For contractility experiments we used diaphragm, soleus (slow-twitch), EDL (fast-twitch) and gastrocnemius (fast-twitch) muscles from wild-type (Wt) and knock-out littermate NebΔExon55

mice. The number of mice used is summarized in Table 1. Muscles were harvested at day 6, as NebΔExon55 mice die one week after

birth22.

Permeabilized muscle fiber contractility

(8)

modifications22. Small fiber bundles (diameter ~70μm) were dissected from the triton-X permeabilized muscles, and attached to aluminum foil clips.

Diaphragm Soleus EDL Gastrocnemius

Wt 6 6 7 6

NebΔExon55 9 6 9 5

Table 1. Number of animals used, for each muscle-type, in Wt and NebΔExon55 mice

The clips were attached to a force transducer (model 403A, Aurora Scientific, Ontario, Canada) and length controller (model 315-CI, Aurora Scientific, Ontario, Canada). We used a setup (model 802D, Aurora Scientific) that was mounted on top of an inverted microscope (Zeiss Axio Observer A1, Zeiss, Thornwood, NY, USA). Sarcomere length was determined using a fast Fourier transformation on a region of interest on the real-time camera image using ASI 900B software (Aurora Scientific Inc., Ontario, Canada).

Experiments were performed at sarcomere length 2.3μm. The width and depth of the muscle bundle was measured with an 40x objective. The cross-sectional area (CSA) was calculated from the average of three consecutive measurements made along the length of the muscle bundle. The temperature was kept constant at 20°C using a TEC controller (ASI 825A, Aurora Scientific Inc. Ontario, Canada).

Various bathing solutions were used during the experimental protocols: a relaxing solution (pCa 9.0), a pre-activating solution with low EGTA concentration and several incremental Ca2+ solutions (pCa 7.0 to 4.5). Composition of these solutions has been described previously24.

The preparations were activated at pCa 4.5 to obtain maximal Ca2+-activated force. Maximal active tension was obtained by divided the force generated at pCa 4.5 by CSA.

(9)

Cross-bridge cycling kinetics (Ktr)

Muscle bundles were first isometrically activated at pCa 4.5, and when a steady tension was reached, cross-bridges were disengaged by performing a quick release of 30% of the initial length, which reduced tension to zero. This was followed by unloaded shortening lasting 30ms. The remaining bound cross-bridges were mechanically detached by rapidly (1ms) re-stretching the muscle fiber bundles to its original length, after which tension redevelops. The Ktr was determined by fitting a double exponential through the force redevelopment curve (note that only the fast rate constant is reported as this is considered to reflect cross-bridge cycling kinetics).

Active stiffness

Following the Ktr protocol, active stiffness was determined by length variations of 0.3, 0.6 and 0.9% on the fiber, resulting a in a force response. Stiffness is represented as the slope of the linear regression of the relationship between tension and length. A typical force trace, including the force response during the Ktr and active stiffness protocol, is shown in Figure 1.

Calcium sensitivity of force generation

Preparations were in relaxing solution and exposed to pre-activating solution (pCa 9.0). Then, they were activated with solutions with a pCa ranging from 7.0 to 4.5 (Figure 2A). The obtained force-pCa relation was fitted with a Hill equation, providing pCa50 (pCa giving 50% of maximal active tension) and the Hill coefficient, nH, an index of myofilament cooperativity (Figure 2B).

Fiber typing

(10)

Figure 1. Example of a force and length trace of a permeabilized fiber bundle activated with exogenous calcium.

The fiber bundle is exposed to saturating calcium concentration to determine the maximal force (Fmax). Second, the fiber is shortened by 30% of the initial length (Lo) to determine the rate constant of force redevelopment (Ktr, red dashed line shows the 2nd order exponential fit from which K

tr is determined). Finally, the

fiber bundle is exposed to length variations of 0.3, 0.6 and 0.9% (blue line) to determine the number of attached cross-bridges.

Muscles fibers were de-natured by boiling at 80C for 2 min in SDS sample buffer. The stacking gel contained a 4% acrylamide concentration (pH 6.7), and the separating gel contained 7% acryl-amide (pH 8.7) with 30% glycerol (v/v). The gels were run for 24 h at 15°C and a constant voltage of 275 V. Finally, the gels were silver-stained, scanned25, and analyzed with AIDA Image Analyzer software (Raytest Isotopenmessgeräte GmbH, Straubenhardt, Gemany).

Statistical Analyses

Data are presented as mean±SEM and % of the Wt. GraphPad Prism 6 was used to generate statistics. For statistical analyses t-test (Wt vs. NebΔexon55 for maximal active tension, K

tr, active stiffness, nH and pCa50) or one-way ANOVA (% of Wt between NebΔexon55) were

performed. P<0.05 was considered to be statistically significant.

(11)

Figure 2. (A) Typical force trace representing the force response to incremental calcium concentrations, and (B) the corresponding force-pCa relation.

The calcium concentration required for half-maximal activation (pCa50) was determined by fitting a modified Hill equation thought the data point (blue line shows the fit from which Hill coefficient (nH) is determined.

RESULTS

The results are shown in Table 2. Maximal active tension (force produced normalized to the cross-sectional area) of diaphragm fibers was reduced by 76% in NebΔexon55 compared to Wt mice

(22.7±2.9 vs. 84.3±2.87 mN/mm2, p<0.0001, respectively). This reduction was significantly larger (p<0.05) than the one observed in NebΔexon55 soleus muscle (reduction of 60%, 21.8±1.4 vs.

53.4±3.16 mN/mm2, p<0.0001, respectively) but not significantly larger than that in NebΔExon55 EDL (reduction of 66%, 19.8±1.2 vs.

60.6±3.8 mN/mm2, p<0.0001, respectively) and gastrocnemius muscles (reduction of 70%, 10.1±1.3 vs. 33.2±6.0 mN/mm2, p=0.0074, respectively).

(12)

Diaphragm Soleus EDL Gastrocnemius Wt

(n=30) Neb

ΔExon55

(n=46) Wt NebΔExon55 Wt NebΔExon55 Wt NebΔExon55 Maximal Active Tension (mN/mm2) (% of Wt) 1.00±0.03 0.27±0.03# 1.00±0.06 0.41±0.02 1.00±0.06 0.33± 0.02 1.00±0.18 0.32±0.04 Ktr (s-1) (% of Wt) 1.00±0.09 0.47±0.08 #& 1.00± 0.04 0.62±0.05 1.00± 0.02 0.63± 0.02 1.00± 0.04 0.50±0.06 Active Stiffness (mN/mm2) (% of Wt) 1.00±0.06 0.25±0.06 1.00±0.13 0.35±0.14 1.00±0.04 0.28±0.03 1.00±0.03 0.27±0.03 Tension/ Stiffness ratio 3.6±0.25 3.1±0.21 3.6±0.3 3.0±0.08 3.3±0.26 2.8±0.2 3.2±0.32 2.9±0.31 pCa50 5.60±0.01 5.50±0.02* 5.86±0.05 5.71±0.05* 5.66±0.04 5.50±0.05* 6.07±0.05 5.91±0.02* nH 4.62±0.42 3.66±0.32 1.65±0.17 1.20±0.10 2.22±0.17 1.71±0.17 3.11±0.44 3.08±0.63 n: fiber number; *p<0.05 Wt vs. NebΔExon55; #

p<0.05 DIA NebΔExon55 vs. SOL NebΔExon55; &

p<0.05 DIA NebΔExon55 vs. EDL NebΔExon55

Table 2. Contractility of permeabilized muscle bundles from Wt and NebΔExon55 mice

gastrocnemius (7.0±0.7 s-1, p<0.0001) NebΔExon55 muscles compared to

the Wt muscles (17.6±1.6, 12.9±1.0, 17.4±0.8, 14.5±0.7, s-1, respectively), with the reduction most pronounced in the diaphragm (Table 2, p<0.05).

To investigate whether the changes in cross-bridge cycling kinetics reduced the number of attached cross-bridges, we measured the active stiffness of the fibers by imposing rapid changes in muscles length. We observed a significant decrease of the active stiffness in NebΔExon55 diaphragm (7.2±1.2 mN/mm2, p<0.0001), soleus (6.3±0.5 mN/mm2, p<0.0001) EDL (6.4±0.6 mN/mm2, p<0.0001) and gastrocnemius (3.0±0.3 mN/mm2, p=0.0014) compared to Wt muscles (28.0±1.7, 17.9±1.1, 22.8±3.0, 11.6±1.7, mN/mm2, respectively), with no differences among muscle types (Table 2).

(13)

We estimated the force generated by cross-bridge by calculating the tension/stiffness ratio. This ratio was not significantly different in NebΔExon55 muscles and Wt and among, muscles types:

diaphragm (3.1±0.21 vs. 3.6±0.25, p=0.2248, respectively), soleus (3.0±0.08 vs. 3.6±0.3, p=0.1078, respectively), EDL (2.8±0.2 vs. 3.3±0.26, p=0.1571, respectively) and gastrocnemius (2.9±0.31 vs. 3.2±0.32, p=0.4670, respectively).

To determine the calcium sensitivity of force generation, we measured force at a range of calcium concentrations from pCa 9.0 to 4.5. A rightward shift of the pCa curve, reflected by a decrease of the pCa50, was observed in NebΔExon55 muscles compare

to Wt (Table 2): diaphragm (5.50±0.02 vs. 5.60±0.01, p=0.0094), soleus (5.71±0.05 vs. 5.86±0.05, p=0.022), EDL (5.50±0.05 vs. 5.66±0.04, p=0.038) and gastrocnemius (5.90±0.03 vs 6.07±0.05, p=0.032) (Table. 2), with no difference among muscle types. This shift indicates that the calcium sensitivity of force is significantly reduced in the NebΔExon55muscles compare to Wt.

The Hill coefficient, representing the cooperativity of activation, is not affected in NebΔExon55 diaphragm (3.66±0.32, p=0.092),

soleus (1.20±0.05, p=0.050), EDL (1.71±0.17, p=0.051) and gastrocnemius (3.01±0.67, p=0.7934) compared to Wt muscles (4.60±0.42, 1.65±0.17, 2.22±0.17, 3.11±0.44, respectively), with no differences among muscle types.

As contractility depends on the MyHC isoforms expressed in the muscle fibers, we analyzed the composition of MyHC isoforms in Wt muscle and NebΔExon55 muscles. Mouse muscle is composed of

(14)

We observed that Wt muscles are composed of a majority of type 2B, followed by type 2A(X) and type 1 (Table 3B). In the

NebΔexon55 diaphragm we observed a decrease of 18% of type 2B

while type 2A(X) increased by 20% compared to Wt. NebΔExon55

EDL presented a similar pattern with a reduction of 17% of type 2B isoform while type 2A(X) increased by 22%. On the other hand, soleus and gastrocnemius muscles did not present any change in MyHC isoform composition. These findings indicate that a change towards slow-type MHC isoforms is not likely to explain the force deficit in NebΔexon55 muscle. (Note that previous work on

permeabilized fibers of slow-twitch soleus and fast-twitch EDL muscle showed comparable maximal tension26, further suggesting that a difference in fiber type does not contribute to the reduced contractility of NebΔexon55 muscle).

DISCUSSION

In the present study, we investigated the effect of deletion of exon 55 in the nebulin gene on the contractility of diaphragm muscle fibers. Diaphragm muscle fibers of NebΔexon55 mice showed severe

contractile weakness, a weakness that was more pronounced than that observed in soleus muscle, but not more pronounced than that in EDL and gastrocnemius muscles. The severe diaphragm weakness likely contributes to the development of respiratory failure in NebΔexon55 mice and might explain their early, postnatal

death22.

Diaphragm weakness in nemaline myopathy

The diaphragm is the main muscle of inspiration, taking up approximately seventy percent of the work of breathing in humans27. Contraction of diaphragm muscle fibers creates a negative thoracic pressure resulting in inspiration. Similar to the heart, the diaphragm is highly active with a duty cycle (ratio of active to inactive time) of ~35%28 compared to 2% for the extensor digitorum longus and 14% for the soleus29. The diaphragm is

(15)

also essential for non-ventilatory behaviors, including expulsive behaviors important for airway clearance. In humans, the diaphragm consists of approximately equal proportions of slow-twitch and fast-slow-twitch muscle fibers30, fibers which are recruited in an orderly fashion with the slow-twitch fibers being recruited first and the fast-twitch fibers mainly during conditions that require high diaphragm force (e.g. during respiratory failure)31. Respiratory failure is the main cause of death in NM, and inspiratory muscle weakness is considered the main contributor to respiratory failure32. Although it is generally accepted that the diaphragm is severely affected in NM, perhaps even much more than peripheral muscles32-35 data on diaphragm contractility in NM patients is scarce and parallel comparison of diaphragm and peripheral muscle strength is, to our knowledge, lacking. Measurements of inspiratory muscle function in NM have been documented in case reports. For instance, Kelly et al.15 reported significantly reduced maximum inspiratory pressures (MIP, a parameter of inspiratory muscle function) in a 44-year old NM-patient resulting in respiratory failure, and a NM-NM-patient with severely reduced MIP was weaned from ventilatory support after inspiratory muscle training improved MIP 2-fold36, indicating the presence and significance of inspiratory muscle weakness in NM. Furthermore, inspiratory muscle weakness in NM has been assessed using spirometry, which provides only indirect measures of diaphragm function. Thus, the goal of the present study was to quantify the magnitude of diaphragm weakness in NM, its cellular pathophysiology, and to compare this magnitude to that in peripheral skeletal muscles.

(16)

Table 3. (A) Example of polyacrylamide gel electrophoresis (SDS-PAGE)

with a homogenate of murine EDL and soleus mixed together and muscle bundles

use of a mouse model for nebulin-based NM. Mutations in NEB account for ~50% of all NM cases and deletion of exon 55 is one of the most frequent mutations in NEB reported so far37-39. The NebΔexon55 mice recapitulate the main features of human

NM (including nemaline rods and manifest muscle weakness) with significant sarcomeric dysfunction in tibialis cranialis muscle22. In the present study, we investigated the contractility of permeabilized muscle fibers, a preparation which allows us to assess the functioning of contractile proteins in the absence of potential confounding effects of for instance calcium cycling by the sarcoplasmic reticulum. These experiments were performed at a sarcomere length of 2.3μm. This sarcomere length was chosen because previous work form our group showed that NebΔexon55

muscle has shorter thin filament lengths, and that at this length the overlap of the thin and thick filaments is optimal. Consequently, weakness in NebΔexon55 diaphragm cannot be attributed to shortened

thin filament length. Our findings indicate manifest dysfunction of sarcomeric proteins in the diaphragm muscle of NebΔExon55

mice. Maximal tension was reduced to ~25% of Wt levels, a reduction at least partly caused by changes in the kinetics of bridge cycling, resulting in a lower number of bound cross-bridges. Note that the tension reduction is more pronounced

(17)

at submaximal (in vivo) stimulation as the calcium sensitivity of force was significantly reduced (Figure 3). Considering that transdiaphragm pressure generation (the gold standard for in vivo diaphragm force) during quiet breathing is ~25% of the maximal transdiaphragm pressure generated during supramaximal bilateral phrenic nerve stimulation (data from small rodents40, 41), our data suggest that normal breathing in the NebΔexon55 mice

requires continuous maximal activation of the diaphragm muscle, a condition that is unlikely to be compatible with life and will likely lead to respiratory failure.

Previous studies on biopsies of NM patients proposed that the diaphragm is more affected than peripheral skeletal muscles42, i.e. more nemaline rods were observed in diaphragm than in non-respiratory muscles. The data of the present study does not support this proposition as two out of the three peripheral skeletal muscles tested exhibited a magnitude of weakness that was comparable to that observed in the diaphragm (Table 2). An explanation for this discrepancy might be that diaphragm pathophysiology in

NebΔexon55 mice does not resemble that of patients. Alternatively,

one could argue that although the number of rods was higher in the diaphragm of patients, the contractility of diaphragm muscle fibers in the patients might have been comparable to that of peripheral muscles fibers (indeed, the number of rods does not seem to correlate with disease severity42).

The diaphragm as therapeutic target in NM

(18)

Figure. 3 Curves indicate the absolute force-calcium relation of diaphragm bundles of Wt and NebΔExon55 mice.

Compared to Wt, at physiological calcium concentration (shade region), the absolute force is reduced by 88%. At pCa 4.5 there is a force decrease of 75% in NebΔExon55 mice compared to Wt.

Similarly, another class of so-called fast skeletal troponin activators induced a large increase in force generation in diaphragm muscle fibers of critically ill patients30, illustrating the promise of these compounds.

Limitations of the study

NebΔexon55 mice die within a week after birth, mimicking the severe

congenital form of nemaline myopathy44. The severe diaphragm of respiratory effort, and the day-time ventilatory support was discontinued. Pharmaceutical compounds might be of use as well, more specifically drugs that augment the function of contractile proteins. In vivo administration of levosimendan, a drug that sensitizes troponin to calcium, improved diaphragm contractility in healthy subjects43 by improving neuromechanical efficiency and by reducing the development of fatigue.

(19)

weakness in these mice supports respiratory failure as a cause of death, however, death from insufficient milk intake due to weakness of other muscles (eg. the facial muscle responsible for suckling) cannot be ruled out. We attempted to isolated hyoid muscles from NebΔexon55 mice for analysis of contractility but were

unsuccessful.

In the present study, we measured the contractility of permeabilized muscle fibers, a preparation in which only the contractile proteins remain functional. This preparation was used because NM is known to specifically affect the functioning of contractile proteins,22, 45 and because measuring the functioning of intact muscle preparations from the diaphragm of six day old mice is challenging and to our knowledge unprecedented. Consequently, the contribution to weakness of upstream processes in excitation-contraction coupling, such as sarcoplasmic reticulum calcium handling, cannot be ruled out, and the 75% reduction in force in permeabilized diaphragm fibers might be an underestimation of the reduction in intact diaphragm muscle.

Typically, patients with NM have a predominance of type 1 muscle fibers, a feature that was not apparent in the muscles that were studied in the NebΔexon55 mice. We speculate that this discrepancy

is a result of the young age at which the NebΔexon55 mice were

studied: the time required for the fiber type shift to occur was too short. Indeed, adult mice with nebulin deficiency do show a predominance of type 1 muscle fibers26.

(20)

material. Note that based on visual observation during the fiber preparation, we observed no differences in connective tissue within muscle types between Wt and NebΔexon55 mice genotypes.

CONCLUSION

Contractile weakness of the diaphragm is prominent in NebΔExon55

mice, but is not more pronounced than that of most peripheral muscle types. Dysfunction of contractile proteins contributes to diaphragm weakness in NebΔexon55 mice, and provides a therapeutic

target to alleviate inspiratory muscle weakness in NM and combat respiratory failure.

(21)

REFERENCES

1. Shy GM, Engel WK, Somers

JE, Wanko T. Nemaline Myopathy. A New Congenital  Myopathy. Brain. 1963 Dec;86:793-810.

2. Nowak KJ, Wattanasirichaigoon D, Goebel HH, et al. Mutations in the skeletal muscle alpha-actin gene in patients with actin myopathy and nemaline myopathy. Nature Genetics. 1999 Oct;23(2):208-12. 3. Laing NG, Wilton SD, Akkari PA, et al. A mutation in the alpha tropomyosin gene TPM3 associated with autosomal dominant nemaline myopathy NEM1. Nat Genet. 1995 Jun;10(2):249.

4. Donner K, Ollikainen M, Ridanpaa M, et al. Mutations in the beta-tropomyosin (TPM2) gene--a rare cause of nemaline myopathy. Neuromuscul Disord. 2002 Feb;12(2):151-8.

5. Pelin K, Hilpela P, Donner K, et al. Mutations in the nebulin gene associated with autosomal recessive nemaline myopathy. Proc Natl Acad Sci U S A. 1999 Mar;96(5):2305-10.

6. Yuen M, Sandaradura SA, Dowling JJ, et al. Leiomodin-3 dysfunction results in thin filament disorganization and nemaline myopathy. The Journal of Clinical Investigation. 2014 Nov;124(11):4693-708. 7. Johnston JJ, Kelley RI, Crawford TO, et al. A novel nemaline myopathy in the Amish caused by a mutation in troponin T1. Am J Hum Genet. 2000 Oct;67(4):814-21.

8. Thirion C, Stucka R, Mendel

B, et al. Characterization of human muscle type cofilin (CFL2) in normal and regenerating muscle. Eur J Biochem. 2001 Jun;268(12):3473-82.

9. Ravenscroft G, Miyatake S,

Lehtokari V-L, et al. Mutations in KLHL40 are a frequent cause of severe

autosomal-recessive nemaline myopathy.

American Journal of Human Genetics. 2013 Jul;93(1):6-18.

10. Gupta VA, Beggs AH. Kelch

proteins: emerging roles in skeletal muscle development and diseases. Skeletal Muscle. 2014 Jun;4:11.

11. Gupta VA, Ravenscroft G, Shaheen R, et al. Identification of KLHL41 Mutations Implicates BTB-Kelch-Mediated Ubiquitination as an Alternate Pathway to Myofibrillar Disruption in Nemaline Myopathy. American journal of human genetics. 2013 Dec;93(6):1108-17.

12. Sambuughin N, Yau KS, Olivé M, et al. Dominant mutations in KBTBD13, a member of the BTB/Kelch family, cause nemaline myopathy with cores. American Journal of Human Genetics. 2010 Dec;87(6):842-7.

13. Malfatti E, Böhm J, Lacène E,

Romero N, Laporte J. A premature stop codon in MYO18B is associated with severe nemaline myopathy with cardiomyopathy. Journal of Neuromuscular Diseases. 2015 Sept;2(3):219-27.

14. Falgà-Tirado C, Pérez-Pemán P, Ordi-Ros J, Bofill JM, Balcells E. Adult onset of nemaline myopathy presenting as respiratory insufficiency. Respiration; International Review of Thoracic Diseases. 1995 Jul;62(6):353-4.

15. Kelly E, Farrell MA, McElvaney NG. Adult-onset nemaline myopathy presenting as respiratory failure. Respiratory Care. 2008 Nov;53(11):1490-4.

16. Whitaker J, Love S, Williams AP, Plummeridge M. Idiopathic adult-onset nemaline myopathy presenting with isolated respiratory failure. Muscle & Nerve. 2009 Mar;39(3):406-8.

17. Maeda MH, Ohta H, Izutsu K,

(22)

nemaline myopathy as a rare cause of slowly progressive muscle weakness with young adult onset. Muscle & Nerve. 2014 Nov; 51:772-774.

18. Taglia A, D'Ambrosio P, Palladino A, Politano L. On a case of respiratory failure due to diaphragmatic paralysis and dilated cardiomyopathy in a patient with nemaline myopathy. Acta Myologica: Myopathies and Cardiomyopathies: Official Journal of the Mediterranean Society of Myology / Edited by the Gaetano Conte Academy for the Study of Striated Muscle Diseases. 2012 Dec;31(3):201-3.

19. Jungbluth H, Sewry CA, Brown SC, et al. Mild phenotype of nemaline myopathy with sleep hypoventilation due to a mutation in the skeletal muscle alpha-actin (ACTA1) gene. Neuromuscular disorders: NMD. 2001 Jan;11(1):35-40.

20. Shahrizaila N, Kinnear WJM, Wills AJ. Respiratory involvement in inherited primary muscle conditions. J Neurol Neurosurg Psychiatry. 2006 Oct;77(10):1108-15.

21. McCool FD, Tzelepis GE. Dysfunction of the diaphragm. N Engl J Med. 2012 Mar;366(10):932-42.

22. Ottenheijm CAC, Buck D, Winter JMd, et al. Deleting exon 55 from the nebulin gene induces severe muscle weakness in a mouse model for nemaline myopathy. Brain. 2013 May;136((Pt 6)):1718-31.

23. Romero NB, Sandaradura SA,

Clarke NF. Recent advances in nemaline myopathy. Current opinion in neurology. 2013 Oct;26(5):519-26.

24. de Winter JM, Joureau B, Sequeira V, et al. Effect of levosimendan on the contractility of muscle fibers from nemaline myopathy patients with mutations in the nebulin gene. Skeletal Muscle. 2015 Apr;5:12. 25. Oakley BR, Kirsch DR, Morris NR.

A simplified ultrasensitive silver stain for detecting proteins in polyacrylamide gels. Analytical Biochemistry. 1980 Jul;105(2):361-3.

26. Li F, Buck D, De Winter J, et al. Nebulin deficiency in adult muscle causes sarcomere defects and muscle-type-dependent changes in trophicity: novel insights in nemaline myopathy. Hum Mol Genet. 2015 Sep;24(18):5219-33.

27. Chandra M, Mamidi R, Ford S, et al. Nebulin alters cross-bridge cycling kinetics and increases thin filament activation: a novel mechanism for increasing tension and reducing tension cost. J Biol Chem. 2009 Nov;284(45):30889-96.

28. Kong FJ, Berger AJ. Firing properties and hypercapnic responses of single phrenic motor axons in the rat. J Appl Physiol (1985). 1986 Dec;61(6):1999-2004.

29. Hensbergen E, Kernell D. Daily durations of spontaneous activity in cat's ankle muscles. Exp Brain Res. 1997 Jun;115(2):325-32.

30. Hooijman PE, Beishuizen A, Witt CC, et al. Diaphragm muscle fiber weakness and ubiquitin-proteasome activation in critically ill patients. Am J Respir Crit Care Med. 2015 May;191(10):1126-38.

31. Sieck GC, Fournier M, Enad JG. Fiber type composition of muscle units in the cat diaphragm. Neurosci Lett. 1989 Feb;97(1-2):29-34.

32. Wallgren-Pettersson C, Sewry CA, Nowak KJ, Laing NG. Nemaline myopathies. Seminars in pediatric neurology. 2011 Dec;18(4):230-8.

33. Tsujihata M, Shimomura C, Yoshimura T, et al. Fatal neonatal nemaline myopathy: a case report. J Neurol Neurosurg Psychiatry. 1983 Sep;46(9):856-9.

34. Matsuo T, Tashiro T, Ikeda T,

Tsujihata M, Shimomura C. Fatal neonatal

(23)

nemaline myopathy. Acta Pathol Jpn. 1982 Sep;32(5):907-16.

35. Gillies C, Raye J, Vasan U, Hart WE, Goldblatt PJ. Nemaline (rod) myopathy: a possible cause of rapidly fatal infantile hypotonia. Arch Pathol Lab Med. 1979 Jan;103(1):1-5.

36. Smith BK, Bleiweis MS, Zauhar J, Martin AD. Inspiratory muscle training in a child with nemaline myopathy and organ transplantation. Pediatr Crit Care Med. 2011 Mar;12(2):e94-8.

37. Lehtokari V-L, Greenleaf RS, DeChene ET, et al. The exon 55 deletion in the nebulin gene--one single founder mutation with world-wide occurrence. Neuromuscular disorders: NMD. 2009 Mar;19(3):179-81. 38. Lehtokari VL, Pelin K, Sandbacka M, et al. Identification of 45 novel mutations in the nebulin gene associated with autosomal recessive nemaline myopathy. Hum Mutat. 2006 Sep;27(9):946-56.

39. Anderson SL, Ekstein J, Donnelly MC, et al. Nemaline myopathy in the Ashkenazi Jewish population is caused by a deletion in the nebulin gene. Hum Genet. 2004 Aug;115(3):185-90.

40. Mantilla CB, Seven YB, Zhan WZ, Sieck GC. Diaphragm motor unit recruitment in rats. Respir Physiol Neurobiol. 2010 Aug;173(1):101-6.

41. Sieck GC. Physiological effects of diaphragm muscle denervation and disuse. Clin Chest Med. 1994 Dec;15(4):641-59. 42. Ryan MM, Ilkovski B, Strickland CD, et al. Clinical course correlates poorly with muscle pathology in nemaline myopathy. Neurology. 2003 Feb;60(4):665-73.

43. Doorduin J, Sinderby CA, Beck J, et al. The calcium sensitizer levosimendan improves human diaphragm function. Am J Respir Crit Care Med. 2012 Jan;185(1):90-5.

44. Saunier M, Bonnemann CG, Durbeej M, Allamand V. 212th ENMC International Workshop:: Animal models of congenital muscular dystrophies, Naarden, The Netherlands, 29-31 May 2015. Neuromuscul Disord. 2016 Feb.

45. Ottenheijm CAC, Hooijman P,

(24)
(25)

Referenties

GERELATEERDE DOCUMENTEN

The moderator that will be added is the self-assessment feeling ‘pride’, and the hypothesis that will be tested is the aim to prove that pride will moderate the effect of life

In additional ANOVA analyses, containing the variable General health interest, it turned out that Assortment structure did have a significant positive direct effect on the

Het blijkt dat beslissers uit hoop op vermij- ding van dit verlies risicozoekend worden en een gunstige regeling die zekerheid biedt, ( ‘neem je beperkte verlies’) af- wijzen in

Publisher’s PDF, also known as Version of Record (includes final page, issue and volume numbers) Please check the document version of this publication:.. • A submitted manuscript is

Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of

1) Teken een lijn als drager van AB en kies daarop een willekeurig punt

Bij planten die werden bijgemest met 0.02 mmol P/l stond de groei na twee weken de groei min of meer stil wanneer geen fosfaat in de voorraad bemesting (?) aanwezig was (tabel