#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

The Polyproline Site in Hinge 2 Influences the Functional Capacity of Truncated Dystrophins


Mutations in dystrophin can lead to Duchenne muscular dystrophy or the more mild form of the disease, Becker muscular dystrophy. The hinge 3 region in the rod domain of dystrophin is particularly prone to deletion mutations. In-frame deletions of hinge 3 are predicted to lead to BMD, however the severity of disease can vary considerably. Here we performed extensive structure-function analyses of truncated dystrophins with modified hinges and spectrin-like repeats in mdx mice. We found that the polyproline site in hinge 2 profoundly influences the functional capacity of a microdystrophinΔR4-R23/ΔCT with a large deletion in the hinge 3 region. Inclusion of polyproline in microdystrophinΔR4-R23/ΔCT led to small myofibers (12% smaller than wild-type), Achilles myotendinous disruption, ringed fibers, and aberrant neuromuscular junctions in the mdx gastrocnemius muscles. Replacing hinge 2 of microdystrophinΔR4-R23/ΔCT with hinge 3 significantly improved the functional capacity to prevent muscle degeneration, increase muscle fiber area, and maintain the junctions. We conclude that the rigid α-helical structure of the polyproline site significantly impairs the functional capacity of truncated dystrophins to maintain appropriate connections between the cytoskeleton and extracellular matrix.


Published in the journal: . PLoS Genet 6(5): e32767. doi:10.1371/journal.pgen.1000958
Category: Research Article
doi: https://doi.org/10.1371/journal.pgen.1000958

Summary

Mutations in dystrophin can lead to Duchenne muscular dystrophy or the more mild form of the disease, Becker muscular dystrophy. The hinge 3 region in the rod domain of dystrophin is particularly prone to deletion mutations. In-frame deletions of hinge 3 are predicted to lead to BMD, however the severity of disease can vary considerably. Here we performed extensive structure-function analyses of truncated dystrophins with modified hinges and spectrin-like repeats in mdx mice. We found that the polyproline site in hinge 2 profoundly influences the functional capacity of a microdystrophinΔR4-R23/ΔCT with a large deletion in the hinge 3 region. Inclusion of polyproline in microdystrophinΔR4-R23/ΔCT led to small myofibers (12% smaller than wild-type), Achilles myotendinous disruption, ringed fibers, and aberrant neuromuscular junctions in the mdx gastrocnemius muscles. Replacing hinge 2 of microdystrophinΔR4-R23/ΔCT with hinge 3 significantly improved the functional capacity to prevent muscle degeneration, increase muscle fiber area, and maintain the junctions. We conclude that the rigid α-helical structure of the polyproline site significantly impairs the functional capacity of truncated dystrophins to maintain appropriate connections between the cytoskeleton and extracellular matrix.

Introduction

Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive disease caused by mutations in the 2.2 MB dystrophin gene [1][3]. In skeletal muscle, dystrophin provides a flexible connection between the cytoskeleton and the dystrophin-glycoprotein complex at the sarcolemma, myotendinous junction (MTJ) and neuromuscular junction (NMJ) [4][6]. Mutations that affect the mechanical integrity of this molecular scaffold render muscles more susceptible to contraction-induced injury leading to cycles of necrosis and regeneration [3].

As a general rule, most frame-shift mutations in dystrophin lead to DMD whereas internal truncations (in-frame deletions) lead to a milder form of the disease called Becker muscular dystrophy (BMD) [7][14]. The severity of BMD can also vary depending on whether a critical region of dystrophin is deleted and the amount of dystrophin being expressed [7][14]. Dystrophin consists of a N-terminal actin-binding domain, a large central rod domain, a cysteine rich region and a C-terminal domain (Figure 1A) [15], [16]. The central rod domain contains 24 spectrin-like repeats, 4 hinges and a second actin-binding domain [15][20]. The locus encoding the N-terminal actin-binding domain and the region near hinge 3 of dystrophin are more susceptible to deletion mutations [7][13]. In-frame deletions of the central rod domain typically lead to a mild BMD [8][13]. However, in-frame deletions at the “hot spot” near hinge 3 can lead to more variable phenotypes [8][13], [21].

The hinge domains of dystrophin influence muscle maturation and maintenance.
Fig. 1. The hinge domains of dystrophin influence muscle maturation and maintenance.
(A) The molecular structure of truncated dystrophins (reviewed in: [15]). ABD1 at the N-terminus is composed of two calponin homology domains denoted by the two circles. The central rod domain contains 24 spectrin-like repeats (R1-24), 4 hinge domains, a 20 amino acid insertion between spectrin-like repeats 15 and 16, and a central actin-binding domain (ABD2). A cluster of basic repeats forms ABD2 that bind to actin through an electrostatic interaction [20]. The hinge domains vary in that hinge 2 contains a polyproline site and hinge 4 contains a WW motif that is required for binding to β-dystroglycan [19], [68]. The cysteine-rich region contains two EF hands and a ZZ domain that is also required for binding to β-dystroglycan. The microdystrophins used in this study are shown below the full-length dystrophin. MicrodystrophinΔR4-R23/ΔCT lacks a large portion of the central rod domain between spectrin-like repeats 4 and 23 and also lacks the C-terminal domain (ΔR4-R23/ΔCT) [24]. Note that microdystrophinΔR4-R23/ΔCT and microdystrophinΔH2-R23+H3/ΔCT differ by a single hinge domain. (B) Transverse sections of gastrocnemius muscles from wild-type, mdx and mdx mice expressing either microdystrophinΔR4-R23/ΔCT or microdystrophinΔH2-R23+H3/ΔCT. Scale bar  = 100 µm. (C) Expression of truncated dystrophins in treated mdx gastrocnemius muscles was similar. The western blots were performed using frozen muscles extracted from OCT. All lanes were loaded equally as shown by α-sarcomeric actin. (D) Shown is the mean +/− S.D. percentage of dystrophin-positive muscle fibers. TA is tibialis anterior. (E) Shown is the mean +/− S.D. of the percentage of muscle fibers containing centrally-located nuclei. MicrodystrophinΔH2-R23+H3/ΔCT with hinge 3 was more effective at preventing muscle degeneration in mdx mice compared to microdystrophinΔR4-R23/ΔCT with hinge 2. The treated muscles show the mean +/− SD for dystrophin-positive fibers only. ***P<0.001 compared to untreated mdx mice. #P<0.05 compared to microdystrophinΔR4-R23/ΔCT treated mdx muscles. (F) Hinge regions of dystrophin influence muscle fiber cross-sectional area. Shown is the mean +/− distribution (25th and 75th percentile (box) in addition to the farthest (whiskers) area of muscle fibers. *P<0.05 and ***P<0.001 compared to wild-type.

The role of dystrophin in vivo has been largely defined by the structure-function relationship of truncated dystrophins in humans and mice [8][13], [22][24]. Rational design of dystrophin mini-genes has been highly effective in preventing and reversing functional abnormalities of dystrophic muscles [22][29]. In particular, we previously developed a microdystrophin (ΔR4-R23/ΔCT; defined as those with 4 or fewer spectrin-like repeats [24]) that accommodates the limited cloning capacity of recombinant adeno-associated viral vectors (rAAV) [24]. Intravenous injection of rAAV vectors pseudotyped with serotype 6 capsid (rAAV6) expressing microdystrophinΔR4-R23/ΔCT can prevent and reverse most aspects of dystrophic pathology in mdx muscles [24], [28], [30][35]. MicrodystrophinΔR4-R23/ΔCT also significantly protects muscles from contraction-induced injury [24], [28], [30][35].

While the microdystrophinΔR4-R23 transgene provides a clear benefit to dystrophic muscles [24], more detailed analyses have revealed a potentially serious abnormality in some muscle groups. The microdystrophinΔR4-R23/mdx transgenic mice have chronic Achilles myotendinous strain injury, which leads to the formation of ringed fibers and fragmentation of the neuromuscular junctions [33], [36]. In the present study we examined whether the domain composition or the small size of microdystrophinΔR4-R23/ΔCT led to this myopathy in mdx mice. We found that the hinge regions of microdystrophin, rather than its small size can profoundly influence skeletal muscle maintenance, maturation and structure.

Results

Dystrophin hinge domains influence the maintenance and maturation of skeletal muscles

We initially screened several truncated dystrophins and found that inclusion of hinge 2, but not hinge 3 could lead to the structural abnormalities we observed in some muscles of the microdystrophinΔR4-R23 transgenic mice (Text S1; Figures S1, S2, S3). We subsequently compared the efficacy of two microdystrophins that differ only in their inclusion of hinge 2 (microdystrophinΔR4-R23/ΔCT) or hinge 3 (microdystrophinΔH2-R23+H3/ΔCT) (Figure 1A) to examine whether the hinge composition of microdystrophin could influence various aspects of muscle disease.

We administered a sub-optimal dose of 2×1012 vector genomes of a rAAV6 pseudotyped vector expressing either microdystrophinΔR4-R23/ΔCT or microdystrophinΔH2-R23+H3/ΔCT intravenously into 2 week-old mdx4cv mice. We used a sub-optimal dose of rAAV6-microdystrophins so that we could examine whether changing the hinge domain increased or decreased the functional capacity of microdystrophin. Six months after treatment, both microdystrophins were expressed in a similar percentage of gastrocnemius and tibialis anterior (TA) muscle fibers (ranging from approximately 61% to 71%; P = 0.238 when comparing between the microdystrophins; Figure 1B and 1D). Western blots confirmed similar expression levels of truncated dystrophins in treated gastrocnemius muscles (Figure 1C). Both microdystrophins restored dystrophin-associated proteins to the sarcolemma except for nNOS (Text S1; Figure S4). MicrodystrophinΔR4-R23/ΔCT containing hinge 2 significantly prevented muscle degeneration (∼11% central nuclei for treated muscles verse ∼78% for untreated mdx muscles; P<0.001), and limited the fiber area of skeletal muscles (12% smaller than wild-type; P<0.05; Figure 1E), consistent with previous studies [24], [32], [33]. MicrodystrophinΔH2-R23+H3/ΔCT containing hinge 3 was significantly better able to prevent muscle degeneration (1–2% central nuclei; P<0.05 compared to microdystrophinΔR4-R23/ΔCT), and surprisingly increased average muscle fiber cross sectional area (34% larger than wild-type; P<0.001; Figure 1E). Thus, replacing hinge 2 of microdystrophinΔR4-R23/ΔCT with hinge 3 significantly improved its capacity to prevent muscle degeneration and promote skeletal muscle maturation.

Dystrophin hinge domains influence myotendinous junction injury and formation of ringed fibers

The tendon extends deep folds into wild-type skeletal muscles to minimize membrane stress under shear (Figure 2)[37]. Most of the folds in the mdx junctions did not extend as far into the gastrocnemius muscles (Figure 2). rAAV6-microdystrophinΔR4-R23/ΔCT severely disrupted the Achilles myotendinous junctions in mdx mice. Many of the junctional folds were missing and myofibril degeneration was evident (Figure 2). Approximately 17% of the adjoining mdx gastrocnemius muscles had ringed fibers. In contrast, rAAV6- microdystrophinΔH2-R23+H3/ΔCT with hinge 3 retained the normal architecture of the Achilles myotendinous junction and we found no ringed fibers in the adjoining gastrocnemius muscles (Figure 2). Thus, the hinge domains influenced whether microdystrophin was capable of maintaining the myotendinous junction and myofibril structure in mdx gastrocnemius muscles.

The hinge domains of dystrophin influence myotendinous junction structure and ringed fiber formation.
Fig. 2. The hinge domains of dystrophin influence myotendinous junction structure and ringed fiber formation.
Shown are electron microscopy images of longitudinal sections of the Achilles myotendinous junctions in addition to transverse sections of gastrocnemius muscles. Note that expression of microdystrophin disrupted the myotendinous junctions and led to formation of ringed fibers (arrow). Scale bar  = 5 µm for myotendinous junctions and 2 µm for transverse sections of gastrocnemius muscles.

Dystrophin hinge domains influence neuromuscular synapse structure

We also examined neuromuscular synapses in mdx mice treated with rAAV6-microdystrophins. Most neuromuscular synapses in wild-type mice (∼97%) form a continuous tertiary structure as shown by staining whole muscle fibers with α-bungarotoxin (Figure 3A). Neuromuscular synapses in mdx mice begin to fragment temporally coincident with muscle degeneration [38]. Approximately 89% of neuromuscular synapses were fragmented in the gastrocnemius muscles of mdx mice (Figure 3B). We had previously shown that the neuromuscular synapses in transgenic microdystrophinΔR4-R23/mdx gastrocnemius muscles fragmented temporally coincident with the formation of ringed fibers [36]. In the present study we found that rAAV6- microdystrophinΔR4-R23/ΔCT containing hinge 2 maintained continuous synapses in only 46% of the mdx gastrocnemius muscles (Figure 3A and 3B). In contrast, approximately 84% of synapses were continuous in mdx gastrocnemius muscles treated with rAAV6-microdystrophinΔH2-R23+H3/ΔCT containing hinge 3 (Figure 3A and 3B).

The hinge domains of dystrophin influence the structure of neuromuscular synapses.
Fig. 3. The hinge domains of dystrophin influence the structure of neuromuscular synapses.
(A) Topographic view of AChR clusters stained with α-bungarotoxin. Scale bar  = 10 µm. (B) Graph shows mean +/− SD percentage of continuous synapses. Significant difference to mdx ***P<0.001. Significant difference to microdystrophinΔR4-R23/ΔCT/mdx mice ###P<0.001. (C) Ultrastructure of neuromuscular synapses from wild-type, mdx and mdx gastrocnemius muscles expressing microdystrophinΔR4-R23/ΔCT and microdystrophinΔH2-R23/ΔCT+H3. Note that synaptic folding is reduced in mdx mice and increased in rAAV6-microdystrophinΔR4-R23/ΔCT treated muscles (arrows). Scale bar  = 0.5 µm. (D) The graph shows the mean +/− SD number of folds per µm of postsynaptic membrane juxtaposed to the presynaptic cleft. Significant difference compared to wild-type **P<0.01. (E) The mean +/− SD depth of the folds was significantly increased in mdx muscles treated with rAAV6-microdystrophinΔR4-R23/ΔCT (***P<0.001).

Neuromuscular synapses also contain folds in the postsynaptic membrane that align directly adjacent to vesicle release sites (active zones) in the pre-synaptic nerve terminal (arrows; Figure 3C). The number of synaptic folds in mdx mice was significantly reduced compared to wild-type (P<0.01; Figure 3C and 3D) as previously described [4], [39]. The number of folds was restored in microdystrophinΔR4-R23/ΔCT and microdystrophinΔH2-R23+H3/ΔCT treated muscles (Figure 3C and 3D). The synaptic folds extended significantly further into microdystrophinΔR4-R23/ΔCT treated mdx muscles compared to wild-type muscles (P<0.001; Figure 3C and 3E), as previously described in transgenic microdystrophinΔR4-R23/mdx mice [36]. In contrast, the number and length of synaptic folds in microdystrophinΔH2-R23+H3/ΔCT treated mdx muscles was similar to wild-type (Figure 3C–3E). Thus, microdystrophinΔH2-R23+H3/ΔCT containing hinge 3 can maintain the structure of neuromuscular junctions in mdx muscles.

Mechanical properties of muscles expressing microdystrophins with either hinge 2 or hinge 3

Contraction-induced injury can initiate muscle degeneration in mdx mice [40]. Skeletal muscles from mdx mice have a lower force producing capacity than wild-type muscles and are more susceptible to contraction-induced injury (Figure 4). We found that sub-optimal doses of both rAAV6-microdystrophinΔR4-R23/ΔCT and rAAV6- microdystrophinΔH2-R23+H3/ΔCT maintained the peak force producing capacity of mdx gastrocnemius and tibialis anterior muscles (Figure 4A). Both microdystrophins also significantly improved the specific force (force per cross sectional area of muscle) production in mdx muscles (P<0.05; Figure 4B). The specific force was not restored to wild-type partly because the sub-optimal dose of rAAV6-microdystrophin did not prevent the pseudo hypertrophy normally found in mdx muscles (P = 0.454 when comparing the muscle mass between mdx and treated mdx muscles; one-way ANOVA). Each microdystrophin significantly protected the treated limb muscles from contraction-induced injury (P<0.001; Figure 4C and 4D). However, we found no significant difference between the peak force, specific force or protection from contraction-induced injury when comparing between the two microdystrophins with either hinge 2 or hinge 3.

Microdystrophins significantly improve the mechanical properties of <i>mdx</i> hind limb muscles.
Fig. 4. Microdystrophins significantly improve the mechanical properties of mdx hind limb muscles.
(A) Graph shows the mean +/− S.D. peak force. *P<0.05 and **P<0.01 compared to wild-type. #P<0.05 and ###P<0.001 compared to mdx. (B) Graph shows the mean +/− S.D. specific force. ***P<0.001 compared to wild-type. #P<0.05 and ##P<0.01 compared to mdx. The contractile performance of (C) gastrocnemius muscles and (D) tibialis anterior muscles immediately prior to increasing length changes during maximal force production. Bars represent the mean +/− S.D. percentage of the initial optimal muscle contraction. rAAV6-microdystrophinΔR4-R23/ΔCT treated muscles were significantly (*P<0.05; **P<0.01; ***P<0.001) protected from contraction-induced injury, as were rAAV6-microdystrophinΔH2-R23/ΔCT+H3 (#P<0.05; ##P<0.01; ###P<0.001) treated muscles when compared to mdx mice.

Polyproline in hinge 2 influences the pathology of skeletal muscle fibers

Together, our results suggested that the structural abnormalities observed in some treated mdx muscles could be traced to the presence of hinge 2 within the microdystrophin. We next examined the molecular composition of the hinges to define what was unique about hinge 2. The hinges in dystrophin are defined as such because of the higher concentration of proline residues, which function to limit the continuation of the α-helical coiled-coils of the spectrin-like repeats through the entire length of the dystrophin rod domain [19]. Both hinge 2 and hinge 3 have six proline residues and the lengths of these hinges are similar [19]. We hypothesized that the placement of the prolines most likely results in their different functions [5], [19]. Hinge 2 has 5 consecutive proline residues (polyproline; Figure 5A) whereas the proline residues in hinge 3 are more evenly distributed throughout the hinge [19]. Polyproline residues are thought to have their own defined rigid helical structure [41], [42], and this could affect the functional capacity of microdystrophinΔR4-R23/ΔCT.

Microdystrophin<sup>ΔR4-R23/ΔCT</sup> did not cause structural abnormalities when the polyproline site was deleted from hinge 2.
Fig. 5. MicrodystrophinΔR4-R23/ΔCT did not cause structural abnormalities when the polyproline site was deleted from hinge 2.
(A) The molecular structure of microdystrophins. Below each structure is the amino acid sequence of hinge 2. Note that proline residues are highlighted in red and that microdystrophinΔPolyP/ΔR4-R23/ΔCT lacks the polyproline site. (B) Transverse sections of gastrocnemius muscles from mdx mice expressing microdystrophinΔR4-R23/ΔCT (top panel) or microdystrophinΔPolyP/ΔR4-R23/ΔCT (lower panel). Scale bar  = 100 µm. (C) Expression of truncated dystrophins from treated gastrocnemius muscles. The western blots were performed using tissue sections frozen in OCT. (D) Shown is the mean +/− S.D. percentage of myofibers with centrally-located nuclei. The treated muscles show the mean +/− SD for dystrophin positive fibers only. ***P<0.001 compared to untreated mdx mice. (E) The polyproline region influenced muscle fiber cross sectional area. Shown is the mean +/− distribution (25th and 75th percentile (box) in addition to the farthest (whiskers) of muscle fibers. ***P<0.001 compared to wild-type. ##P<0.01 compared to microdystrophinΔR4-R23/ΔCT. (F) MicrodystrophinΔR4-R23/ΔCT expression led to a disruption of the myotendinous junctions and ringed fiber formation (arrow; top panels) in mdx gastrocnemius muscles, but not when the polyproline site was deleted (lower panels). Scale bars  = 2 µm. (G) Electron microscopy images of neuromuscular junctions (NMJ) in mdx muscles treated with rAAV6-microdystrophinΔR4-R23/ΔCT or rAAV6- microdystrophinΔPolyP/ΔR4-R23/ΔCT. Scale bar  = 1 µm. (H) The graph shows the mean +/− SD number of folds per mm of postsynaptic membrane juxtaposed to the presynaptic cleft. (I) Deletion of the polyproline site from microdystrophinΔR4-R23/ΔCT restored the normal mean +/− SD length of synaptic folds (***P<0.001 compared to microdystrophinΔR4-R23/ΔCT).

To test this hypothesis we compared muscles expressing the original microdystrophinΔR4-R23/ΔCT with a newly developed microdystrophinΔpolyP/ΔR4-R23/ΔCT that lacks the polyproline site in hinge 2 (Figure 5A). We delivered 6×1010 vg of each microdystrophin into mdx gastrocnemius muscles at 2 days of age and examined the mice 7 weeks after treatment. Both microdystrophins were expressed in a similar percentage of muscle fibers (Figure 5B; 59–68%), and were expressed at similar levels (Figure 5C). Each microdystrophin significantly reduced muscle fiber degeneration (Figure 5D). As expected, the original microdystrophinΔR4-R23/ΔCT limited muscle fiber cross-sectional area (Figure 5E), was associated with disrupted myotendinous junctions (Figure 5F), led to the formation of ringed fibers (Figure 5F), and perturbed neuromuscular junctions (Figure 5G–5I). In contrast, the mdx muscles treated with microdystrophinΔpolyP/ΔR4-R23/ΔCT did not show any abnormalities in muscle fiber maturation or structure (Figure 5). Thus, the presence of this polyproline site in hinge 2 of microdystrophinΔR4-R23/ΔCT prevented the appropriate integration of muscles into the nerve-tendon environment.

Discussion

Most gene therapy strategies for DMD require the generation of highly functional truncated dystrophins. rAAV is an efficient and safe vector for systemically delivering truncated dystrophins to striated muscles to prevent muscle degeneration in animal models of DMD ([28]; reviewed in [43]). We had previously generated a microdystrophinΔR4-R23 that was highly capable of mitigating muscle degeneration and improving the mechanical function of mdx skeletal muscles [24], [28]. However, the microdystrophinΔR4-R23 transgene leads to chronic strain injury at the Achilles myotendinous junction [33]. This led to the formation of ringed fibers that function to protect skeletal muscles from contraction-induced injury, even better than wild-type mice [33]. The formation of the rings led to fragmentation of the neuromuscular junctions [36]. Other effects of the transgene included smaller muscle fibers [24], and increased length of synaptic folds [36]. Here we found that each of these phenotypic changes was recapitulated in mdx gastrocnemius muscles treated with rAAV6-microdystrophinΔR4-R23/ΔCT. A screen of several newly developed dystrophin mini-genes revealed that the hinge 2 region influenced the functional capacity of microdystrophinΔR4-R23/ΔCT. Replacing hinge 2 with hinge 3 led to several advantages such as better protection of skeletal muscles (only 1–2% central nuclei 6 months post treatment), larger muscle fibers and normal junctions. Deleting the polyproline site from hinge 2 of microdystrophinΔR4-R23/ΔCT also prevented these structural abnormalities.

Mechanical properties of skeletal muscles expressing microdystrophins

MicrodystrophinΔH2-R23+H3/ΔCT with hinge 3 significantly increased peak force, specific force and protected muscles from contraction-induced injury. However, the morphological improvements of microdystrophinΔH2-R23+H3/ΔCT treated muscles did not translate into a functional improvement compared to microdystrophinΔR4-R23/ΔCT treated muscles. This could result from the molecular and cellular responses to myotendinous strain injury that help protect the rAAV6-microdystrophinΔR4-R23/ΔCT treated muscles from contraction-induced injury [33]. Another possibility is that the presence of some dystrophin negative fibers masked any functional difference between the two proteins. The inclusion of hinge 2 in microdystrophin limited muscle fiber area whereas the inclusion of hinge 3 increased muscle fiber area (Figure 1). Larger muscle fibers in microdystrophinΔH2-R23+H3/ΔCT treated mice could have two distinct advantages: They could replace some of the muscle mass lost in advanced stages of disease and they could be better protected from contraction-induced injury [44]. However, the sub-optimal dose of either rAAV6-microdystrophin did not prevent the pseudo hypertrophy in mdx mice and no mechanical advantages could be discerned when comparing treatments. Saturating levels of rAAV6-microdystrophins or transgenic mice will most likely be required to detect minor differences in the mechanical properties of muscles expressing various truncated dystrophins.

How does polyproline influence the functional capacity of truncated dystrophins?

Our most effective truncated dystrophins developed for gene therapy have been designed to maximize functional interactions between specific spectrin-like repeats and hinge domains. This design has been influenced by genetic studies in mice and man as well as biophysical studies in vitro on the structure, folding and physical properties of both individual and tandemly expressed spectrin-like repeats and hinge domains [24], [45][52]. Individual spectrin-like repeats are not all interchangeable, and ones adjacent to hinges have distinct properties from those flanked by other spectrin-like repeats [21], [24], [47], [51], [52]. Also, spectrin-like repeats rarely function as isolated units [15], [24], [50][53]. Instead, they appear to fold into nested domains interrupted by various insertions (hinges) that disrupt the uniformity and rigidity of the spectrin-like repeat rod domain [24], [45][48], [53][55]. These interruptions appear important for the elastic and flexible structure that dystrophin requires in its role as a force transducer and shock absorber in muscle [56][59]. Our studies suggest that the most functional truncations of dystrophin retain a central hinge domain that is flanked by spectrin-like repeats found adjacent to a hinge in the wild-type dystrophin [24]. Disruption of this linkage could influence protein folding, stability and function leading to the variable phenotypes in patients associated with deletions at or near hinge 3, which is encoded on exons 50–51 [21].

Individual spectrin-like repeats are composed of 3 helical domains connected by non-helical linkers, which fold into a triple helical coiled coil structure (Figure 5A; [45], [47]). The linker regions between discreet repeats are also typically short and relatively unstructured to allow a smooth connection between the third helix of a preceding repeat and the first helix of the next repeat (Figure 6A). However, hinge domains interrupt the nested nature of adjacent spectrin-like repeats and allow more flexibility in the rod domain (Figure 6B). This degree of flexibility appears to be significantly different when hinge 2 or hinge 3 is present. While both hinges contain 6 prolines, which act to disrupt alpha helical structures, in hinge 3 they are dispersed whereas 5 of the 6 prolines in hinge 2 are clustered together (Figure 5A, Figure 6C and 6D; [10]). Polyproline residues form a rigid α-helix [41], [42], much like a molecular ruler [60]. We suggest that the location of this polyproline sequence within a highly truncated rod domain induces a severe structural disruption that can affect the ability of dystrophin to form a mechanically flexible connection between F-actin and β-dystroglycan. Spectrin-like repeats 1-3 have been shown to associate with the sarcolemmal membrane, while the WW domain in hinge 4 forms a critical portion of the β-dystroglycan binding domain [45], [61]. A rigid rod domain induced by polyproline in hinge 2 may directly impair the ability of microdystrophin to form a flexible interaction with either or both of these structures (Figure 6C). In contrast, when hinge 2 is present in full-length dystrophin, a significantly greater number of spectrin-like repeats are present between the hinge and the β-dystroglycan binding domain, allowing greater flexibility in the overall structure.

Predicted nested structure of specific dystrophin spectrin-like repeats relevant to microdystrophins and how they interact with hinge domains.
Fig. 6. Predicted nested structure of specific dystrophin spectrin-like repeats relevant to microdystrophins and how they interact with hinge domains.
(A) Predicted structure of 3 nested repeats [15]. Each repeat is composed of 3 helical domains (a, b, c) connected by non-helical linkers. The triple helical coiled-coil repeat structure is formed by helices b and c of the preceding repeat interfacing with the N-terminal helix a' from the following repeat. The helices a and b fold together into a long repeat, while helix c folds into the short repeat. (B) In dystrophin, the repeat domains are interrupted by hinge 2 (between repeats 3 and 4) and by hinge 3 (Repeats19 and 20). Shown is the predicted structure of the repeat 19-hinge 3-repeat 20 domain in full-length dystrophin. (C) The hinge 2-microdystrophin induces an unusual structural alteration that disrupts the normal connection between adjacent repeats. (D) In contrast, use of hinge 3 generates a similar structure as in (B) except that hinge 3 joins to repeats 3 and 24 rather than repeats 19 and 20.

It is difficult to predict the function of the polyproline site from patients with in frame deletions of exon 17 (hinge 2) of dystrophin. The described deletions (Leiden Muscular Dystrophy Pages) usually encompass larger regions of dystrophin than the polyproline site and it is not clear how these deletions affect protein stability. Our finding that hinge 3 microdystrophin can prevent muscle degeneration suggests that the polyproline site is not a necessary component of dystrophin, similar to previous reports on longer forms of truncated dystrophins [24], [62].

Flanigan et. al., 2009 has proposed that approximately 62% of all DMD patients could be treated with oligonucleotides that skip exons 45–55 (from spectrin-like repeat 18–22)[14]. This would create a truncated dystrophin that contains hinge 2 but not hinge 3, similar to, but much larger than our microdystrophinΔR4-R23 transgene. It will therefore be of interest to determine whether the polyproline site in hinge 2 can influence the functional capacity of larger, truncated dystrophins. It will also be of interest to examine whether the polyproline site affects the functional capacity of truncated utrophin constructs that are designed for gene therapy of DMD [63], [64].

Materials and Methods

Mice and ethics statement

We utilized C57Bl/10 wild-type mice and mdx4cv mice. All experiments are in accordance with the institution of animal care and use committee (IACUC) of the University Of Washington.

Generation of constructs

The expression vector CMV-ΔR4-R23/ΔCT which uses the cytomegalovirus immediate early promoter and enhancer to drive expression of a microdystrophin cDNA was generated as previously described [32]. We generated the ΔH2-R24/ΔCT, ΔR2-R23+R18-H3/ΔCT, ΔH2-R23+H3/ΔCT and ΔPolyP/ΔR4-R23/ΔCT constructs using recombination PCR with CMV-ΔR4-R23/ΔCT as the template [65]. The primers used to generate ΔH2-R24/ΔCT, ΔR2-R23+R18-H3/ΔCT, ΔH2-R23+H3/ΔCT and ΔPolyP/ΔR4-R23/ΔCT are found in Table S1. The resulting expression vectors were sequenced and co-transfected with the pDGM6 packaging plasmid into HEK293 cells to generate recombinant AAV vectors comprising serotype 6 capsids that were harvested, purified, and quantitated as described previously [29]. The resulting titer was determined by comparison to previously known concentrations of rAAV6-CMV-lacZ and ΔR4-R23/ΔCT by Southern analyses with a probe to the CMV promoter. The rAAV6-microdystrophins were delivered intravenously by tail vein injection at two weeks of age or directly into the mdx gastrocnemius muscles at 2 days of age while the mice were anaesthetized.

Gross muscle morphology and morphometry

Gross muscle morphology was analyzed as previously described [24], [32]. Primary antibodies included the N-terminus of dystrophin (1∶800; [23]), utrophin A (1∶300; gift from Stanley Froehner, University of Washington), mouse monoclonal anti-α-dystrobrevin (Transduction laboratories; 1∶200), rabbit polyclonal anti-Syn17 (α-syntrophin; 1∶200; [66]), rabbit polyclonal anti-nNOS (Alexis; 1∶200). Secondary antibodies included Alexa 488, Alexa 594 rabbit polyclonal or Alexa 488 mouse monoclonal secondary antibodies (Molecular Probes; 1∶800). The sections were mounted in anti-fade mounting media containing DAPI (Vector Labs). Fluorescent sections were imaged using a Nikon eclipse E1000 fluorescent microscope (Nikon; NY) and captured using a DeltaVision fluorescence microscope. Muscle fiber areas were quantified using Image J (NIH).

Immunoblotting

For immunoblots, n = 4 gastrocnemius muscles from mdx mice and mdx mice treated with rAAV6-microdystrophinΔR4-R23/ΔCT or rAAV6-microdystrophinΔH2-R23+H3/ΔCT were thawed from OCT blocks and placed into extract buffer (50 mM Tris-HCl, 150 mM NaCl, 0.2% sodium dodecyl sulfate, 10% glycerol, 24 mM Na Deoxycholate, 1% NP40, 47.6 mM Na Fluoride, 200 mM Na orthovanadate, Roche). Protein concentrations were determined by Coomassie Plus Bradford Assay (Peirce). Equal amounts of protein (15 mg) were resolved on a 4–12% SDS polyacrylamide gel. The blots were incubated in rabbit polyclonal antibodies to dystrophin (1∶500; kind gift from James Ervasti, University of Minnesota) and mouse monoclonal antibodies to α-sarcomeric actin (1∶500; SIGMA).

We also performed immunoblots on frozen tissue sections from n = 4 gastrocnemius muscles treated with rAAV6-microdystrophinΔR4-R23/ΔCT and microdystrophinΔPolyP/ΔR4-R23/ΔCT as previously described [67], with minor modifications. Briefly, we cut twenty-five 20 µm sections and diluted the sections into 200 µl lysis buffer (4% SDS, 25 mM Tris pH 8.8, 40% glycerol, 0.5 M phenylmethylsulfonyl fluoride, 100 mM dithiothreitol and bromophenol blue). Samples were briefly sonicated (10 sec at 4°C), heated to 95°C for 5 minutes, centrifuged for 5 minutes at 13,200×g and electrophoresed on a 4–12% SDS-polyacrylamide gel. The blots were incubated in primary rabbit polyclonal antibody against the N-terminus of dystrophin (1∶500; kind gift from James Ervasti, University of Minnesota). All blots were developed with ECL Plus (Pierce) and scanned with the Storm 860 imaging system (Amersham Biosciences).

Electron microscopy

Electron microscopy was performed as previously described [33]. The junctional fold number and lengths were measured from n = 4 mice at 6 months of age using Image J (NIH) and compared using Students t-test (Prism). The counts represent the fold numbers and lengths from all fibers (dystrophin positive and negative).

Quantitation of ringed fibers

We quantitated the number of ringed myofibers in EM images and thick (1 µm) toluidine blue sections from at least 4 animals per group. At least 300 muscle fibers from n = 4 gastrocnemius muscles were examined from wild-type, mdx4cv and mdx4cv mice expressing the various microdystrophins.

Quantification of neuromuscular synapses

Neuromuscular synapses were analyzed in whole mount immunofluorescence stained muscles and quantitated as previously described [36]. The acetylcholine receptor clusters were stained with TRITC conjugated α-bungarotoxin (αBTX; 1∶800; Molecular Probes). Synapses were classified as continuous if they presented with 3 or less continuous regions of AChR clustering and discontinuous if they presented with more than 3 regions of AChR clustering. More than 50 synapses were analyzed from treated and untreated gastrocnemius skeletal muscle fibers from n = 4 mice. The counts in treated muscles include both dystrophin positive and negative fibers. We compared the proportion of continuous synapses using a Students t-test.

Muscle physiology

Muscle physiology was performed as previously described for tibialis anterior [29] and gastrocnemius [33] muscles. We examined six-month-old wild-type, mdx, and mdx mice treated with rAAV6-microdystrophinΔR4-R23/ΔCT or rAAV6-microdystrophinΔH2-R23+H3/ΔCT (n = 5).

Supporting Information

Attachment 1

Attachment 2

Attachment 3

Attachment 4

Attachment 5

Attachment 6


Zdroje

1. HoffmanEP

BrownRHJr

KunkelLM

1987 Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 51 919 928

2. KoenigM

HoffmanEP

BertelsonCJ

MonacoAP

FeenerC

1987 Complete cloning of the Duchenne muscular dystrophy (DMD) cDNA and preliminary genomic organization of the DMD gene in normal and affected individuals. Cell 50 509 517

3. EmeryAE

Muntoni

2003 Duchenne Muscular Dystrophy., 3rd edn. Oxford University Press, Oxford

4. BanksGB

FuhrerC

AdamsME

FroehnerSC

2003 The postsynaptic submembrane machinery at the neuromuscular junction: requirement for rapsyn and the utrophin/dystrophin-associated complex. J Neurocytol 32 709 726

5. BhasinN

LawR

LiaoG

SaferD

EllmerJ

2005 Molecular extensibility of mini-dystrophins and a dystrophin rod construct. J Mol Biol 352 795 806

6. ErvastiJM

2007 Dystrophin, its interactions with other proteins, and implications for muscular dystrophy. Biochim Biophys Acta 1772 108 117

7. ChamberlainJS

GibbsRA

RanierJE

NguyenPN

CaskeyCT

1988 Deletion screening of the Duchenne muscular dystrophy locus via multiplex DNA amplification. Nucleic Acids Res 16 11141 11156

8. BaumbachLL

ChamberlainJS

WardPA

FarwellNJ

CaskeyCT

1989 Molecular and clinical correlations of deletions leading to Duchenne and Becker muscular dystrophies. Neurology 39 465 474

9. GillardEF

ChamberlainJS

MurphyEG

DuffCL

SmithB

1989 Molecular and phenotypic analysis of patients with deletions within the deletion-rich region of the Duchenne muscular dystrophy (DMD) gene. Am J Hum Genet 45 507 520

10. KoenigM

BeggsAH

MoyerM

ScherpfS

HeindrichK

1989 The molecular basis for Duchenne versus Becker muscular dystrophy: correlation of severity with type of deletion. Am J Hum Genet 45 498 506

11. WerneckLC

ScolaRH

MaegawaGH

WerneckMC

2001 Comparative analysis of PCR-deletion detection and immunohistochemistry in Brazilian Duchenne and Becker muscular dystrophy patients. Am J Med Genet 103 115 120

12. Den DunnenJT

GrootscholtenPM

BakkerE

BlondenLA

GinjaarHB

1989 Topography of the Duchenne muscular dystrophy (DMD) gene: FIGE and cDNA analysis of 194 cases reveals 115 deletions and 13 duplications. Am J Hum Genet 45 835 847

13. Coral-VazquezR

ArenasD

CisnerosB

PenalozaL

KofmanS

1993 Analysis of dystrophin gene deletions in patients from the Mexican population with Duchenne/Becker muscular dystrophy. Arch Med Res 24 1 6

14. FlaniganKM

DunnDM

von NiederhausernA

SoltanzadehP

GappmaierE

2009 Mutational spectrum of DMD mutations in dystrophinopathy patients: application of modern diagnostic techniques to a large cohort. Hum Mutat 30 1657 1666

15. AbmayrS

ChamberlainJ

2006 The structure and function of dystrophin.

WinderSJ

Molecular Mechanisms of Muscular Dystrophies Georgetown Landes Biosciences 14 34

16. BanksGB

ChamberlainJS

2008 The value of mammalian models for duchenne muscular dystrophy in developing therapeutic strategies. Curr Top Dev Biol 84 431 453

17. KoenigM

MonacoAP

KunkelLM

1988 The complete sequence of dystrophin predicts a rod-shaped cytoskeletal protein. Cell 53 219 226

18. DavisonMD

BaronMD

CritchleyDR

WoottonJC

1989 Structural analysis of homologous repeated domains in alpha-actinin and spectrin. Int J Biol Macromol 11 81 90

19. KoenigM

KunkelLM

1990 Detailed analysis of the repeat domain of dystrophin reveals four potential hinge segments that may confer flexibility. J Biol Chem 265 4560 4566

20. AmannKJ

RenleyBA

ErvastiJM

1998 A cluster of basic repeats in the dystrophin rod domain binds F-actin through an electrostatic interaction. J Biol Chem 273 28419 28423

21. CarsanaA

FrissoG

TremolaterraMR

LanzilloR

VitaleDF

2005 Analysis of dystrophin gene deletions indicates that the hinge III region of the protein correlates with disease severity. Ann Hum Genet 69 253 259

22. PhelpsSF

HauserMA

ColeNM

RafaelJA

HinkleRT

1995 Expression of full-length and truncated dystrophin mini-genes in transgenic mdx mice. Hum Mol Genet 4 1251 1258

23. RafaelJA

CoxGA

CorradoK

JungD

CampbellKP

1996 Forced expression of dystrophin deletion constructs reveals structure-function correlations. J Cell Biol 134 93 102

24. HarperSQ

HauserMA

DelloRussoC

DuanD

CrawfordRW

2002 Modular flexibility of dystrophin: implications for gene therapy of Duchenne muscular dystrophy. Nat Med 8 253 261

25. CrawfordGE

FaulknerJA

CrosbieRH

CampbellKP

FroehnerSC

2000 Assembly of the dystrophin-associated protein complex does not require the dystrophin COOH-terminal domain. J Cell Biol 150 1399 1410

26. WangB

LiJ

XiaoX

2000 Adeno-associated virus vector carrying human minidystrophin genes effectively ameliorates muscular dystrophy in mdx mouse model. Proc Natl Acad Sci U S A 97 13714 13719

27. YuasaK

MiyagoeY

YamamotoK

NabeshimaY

DicksonG

1998 Effective restoration of dystrophin-associated proteins in vivo by adenovirus-mediated transfer of truncated dystrophin cDNAs. FEBS Lett 425 329 336

28. GregorevicP

BlankinshipMJ

AllenJM

CrawfordRW

MeuseL

2004 Systemic delivery of genes to striated muscles using adeno-associated viral vectors. Nat Med 10 828 834

29. GregorevicP

AllenJM

MinamiE

BlankinshipMJ

HaraguchiM

2006 rAAV6-microdystrophin preserves muscle function and extends lifespan in severely dystrophic mice. Nat Med 12 787 789

30. YoshimuraM

SakamotoM

IkemotoM

MochizukiY

YuasaK

2004 AAV vector-mediated microdystrophin expression in a relatively small percentage of mdx myofibers improved the mdx phenotype. Mol Ther 10 821 828

31. YueY

LiuM

DuanD

2006 C-terminal-truncated microdystrophin recruits dystrobrevin and syntrophin to the dystrophin-associated glycoprotein complex and reduces muscular dystrophy in symptomatic utrophin/dystrophin double-knockout mice. Mol Ther 14 79 87

32. BanksGB

GregorevicP

AllenJM

FinnEE

ChamberlainJS

2007 Functional capacity of dystrophins carrying deletions in the N-terminal actin-binding domain. Hum Mol Genet 16(17) 2105 2113

33. BanksGB

CombsAC

ChamberlainJR

ChamberlainJS

2008 Molecular and cellular adaptations to chronic myotendinous strain injury in mdx mice expressing a truncated dystrophin. Hum Mol Genet (17)24 3975 3986

34. FosterH

SharpPS

AthanasopoulosT

TrolletC

GrahamIR

2008 Codon and mRNA Sequence Optimization of Microdystrophin Transgenes Improves Expression and Physiological Outcome in Dystrophic mdx Mice Following AAV2/8 Gene Transfer. Mol Ther

35. GregorevicP

BlankinshipMJ

AllenJM

ChamberlainJS

2008 Systemic microdystrophin gene delivery improves skeletal muscle structure and function in old dystrophic mdx mice. Mol Ther 16 657 664

36. BanksGB

ChamberlainJS

FroehnerSC

2009 Truncated dystrophins can influence neuromuscular synapse structure. Mol Cell Neurosci 40 433 441

37. TidballJG

1991 Force transmission across muscle cell membranes. J Biomech 24 Suppl 1 43 52

38. LyonsPR

SlaterCR

1991 Structure and function of the neuromuscular junction in young adult mdx mice. J Neurocytol 20 969 981

39. GradyRM

TengH

NicholMC

CunninghamJC

WilkinsonRS

1997 Skeletal and cardiac myopathies in mice lacking utrophin and dystrophin: a model for Duchenne muscular dystrophy. Cell 90 729 738

40. LynchGS

2004 Role of contraction-induced injury in the mechanisms of muscle damage in muscular dystrophy. Clin Exp Pharmacol Physiol 31 557 561

41. KayBK

WilliamsonMP

SudolM

2000 The importance of being proline: the interaction of proline-rich motifs in signaling proteins with their cognate domains. Faseb J 14 231 241

42. RathA

DavidsonAR

DeberCM

2005 The structure of “unstructured” regions in peptides and proteins: role of the polyproline II helix in protein folding and recognition. Biopolymers 80 179 185

43. JudgeLM

ChamberlainJS

2005 Gene therapy for Duchenne muscular dystrophy: AAV leads the way. Acta Myol 24 184 193

44. GehrigSM

KoopmanR

NaimT

TjoakarfaC

LynchGS

2009 Making Fast-Twitch Dystrophic Muscles Bigger Protects Them from Contraction Injury and Attenuates the Dystrophic Pathology. Am J Pathol

45. CrossRA

StewartM

Kendrick-JonesJ

1990 Structural predictions for the central domain of dystrophin. FEBS Lett 262 87 92

46. YanY

WinogradE

VielA

CroninT

HarrisonSC

1993 Crystal structure of the repetitive segments of spectrin. Science 262 2027 2030

47. KahanaE

MarshPJ

HenryAJ

WayM

GratzerWB

1994 conformation and phasing of dystrophin structural repeats. J Mol Biol 235 1271 1277

48. CalvertR

KahanaE

GratzerWB

1996 Stability of the dystrophin rod domain fold: evidence for nested repeating units. Biophys J 71 1605 1610

49. PascualJ

CastresanaJ

SarasteM

1997 Evolution of the spectrin repeat. Bioessays 19 811 817

50. BroderickMJ

WinderSJ

2002 Towards a complete atomic structure of spectrin family proteins. J Struct Biol 137 184 193

51. HarperSQ

CrawfordRW

DelloRussoC

ChamberlainJS

2002 Spectrin-like repeats from dystrophin and alpha-actinin-2 are not functionally interchangeable. Hum Mol Genet 11 1807 1815

52. SaadatL

PittmanL

MenhartN

2006 Structural cooperativity in spectrin type repeats motifs of dystrophin. Biochim Biophys Acta 1764 943 954

53. WinderSJ

GibsonTJ

Kendrick-JonesJ

1995 Dystrophin and utrophin: the missing links! FEBS Lett 369 27 33

54. KahanaE

GratzerWB

1995 Minimum folding unit of dystrophin rod domain. Biochemistry 34 8110 8114

55. PascualJ

PfuhlM

WaltherD

SarasteM

NilgesM

1997 Solution structure of the spectrin repeat: a left-handed antiparallel triple-helical coiled-coil. J Mol Biol 273 740 751

56. ErvastiJM

CampbellKP

1993 A role for the dystrophin-glycoprotein complex as a transmembrane linker between laminin and actin. J Cell Biol 122 809 823

57. RybakovaIN

PatelJR

ErvastiJM

2000 The dystrophin complex forms a mechanically strong link between the sarcolemma and costameric actin. J Cell Biol 150 1209 1214

58. ErvastiJM

2003 Costameres: the Achilles' heel of Herculean muscle. J Biol Chem 278 13591 13594

59. OzawaE

2006 The functional biology of dystrophin: structural components and the pathogenesis of Duchenne muscular dystrophy.

ChamberlainJS

RandoTA

Duchenne Muscular Dystrophy: Advances in Therapeutics New York Taylor and Francis 21 54

60. MoradiM

BabinV

RolandC

DardenTA

SaguiC

2009 Conformations and free energy landscapes of polyproline peptides. Proc Natl Acad Sci U S A

61. Ishikawa-SakuraiM

YoshidaM

ImamuraM

DaviesKE

OzawaE

2004 ZZ domain is essentially required for the physiological binding of dystrophin and utrophin to beta-dystroglycan. Hum Mol Genet 13 693 702

62. LaiY

ThomasGD

YueY

YangHT

LiD

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

63. OdomGL

GregorevicP

AllenJM

FinnE

ChamberlainJS

2008 Microutrophin delivery through rAAV6 increases lifespan and improves muscle function in dystrophic dystrophin/utrophin-deficient mice. Mol Ther 16 1539 1545

64. SonnemannKJ

Heun-JohnsonH

TurnerAJ

BaltgalvisKA

LoweDA

2009 Functional substitution by TAT-utrophin in dystrophin-deficient mice. PLoS Med 6 e1000083 doi:10.1371/journal.pmed.1000083

65. ChamberlainJ

2004 PCR-mediated mutagenesis. In: Nature Encyclopedia of Life Sciences. Nature Publishing Group, London

66. PetersMF

AdamsME

FroehnerSC

1997 Differential association of syntrophin pairs with the dystrophin complex. J Cell Biol 138 81 93

67. CooperST

LoHP

NorthKN

2003 Single section Western blot: improving the molecular diagnosis of the muscular dystrophies. Neurology 61 93 97

68. ErvastiJM

OhlendieckK

KahlSD

GaverMG

CampbellKP

1990 Deficiency of a glycoprotein component of the dystrophin complex in dystrophic muscle. Nature 345 315 319

Štítky
Genetika Reprodukční medicína

Článek vyšel v časopise

PLOS Genetics


2010 Číslo 5
Nejčtenější tento týden
Nejčtenější v tomto čísle
Kurzy

Zvyšte si kvalifikaci online z pohodlí domova

Svět praktické medicíny 1/2024 (znalostní test z časopisu)
nový kurz

Koncepce osteologické péče pro gynekology a praktické lékaře
Autoři: MUDr. František Šenk

Sekvenční léčba schizofrenie
Autoři: MUDr. Jana Hořínková

Hypertenze a hypercholesterolémie – synergický efekt léčby
Autoři: prof. MUDr. Hana Rosolová, DrSc.

Význam metforminu pro „udržitelnou“ terapii diabetu
Autoři: prof. MUDr. Milan Kvapil, CSc., MBA

Všechny kurzy
Kurzy Podcasty Doporučená témata Časopisy
Přihlášení
Zapomenuté heslo

Zadejte e-mailovou adresu, se kterou jste vytvářel(a) účet, budou Vám na ni zaslány informace k nastavení nového hesla.

Přihlášení

Nemáte účet?  Registrujte se

#ADS_BOTTOM_SCRIPTS#