Skip to main content
Erschienen in: BMC Neurology 1/2018

Open Access 01.12.2018 | Research article

LARGE expression in different types of muscular dystrophies other than dystroglycanopathy

verfasst von: Burcu Balci-Hayta, Beril Talim, Gulsev Kale, Pervin Dincer

Erschienen in: BMC Neurology | Ausgabe 1/2018

Abstract

Background

Alpha-dystroglycan (αDG) is an extracellular peripheral glycoprotein that acts as a receptor for both extracellular matrix proteins containing laminin globular domains and certain arenaviruses. An important enzyme, known as Like-acetylglucosaminyltransferase (LARGE), has been shown to transfer repeating units of -glucuronic acid-β1,3-xylose-α1,3- (matriglycan) to αDG that is required for functional receptor as an extracellular matrix protein scaffold. The reduction in the amount of LARGE-dependent matriglycan result in heterogeneous forms of dystroglycanopathy that is associated with hypoglycosylation of αDG and a consequent lack of ligand-binding activity. Our aim was to investigate whether LARGE expression showed correlation with glycosylation of αDG and histopathological parameters in different types of muscular dystrophies, except for dystroglycanopathies.

Methods

The expression level of LARGE and glycosylation status of αDG were examined in skeletal muscle biopsies from 26 patients with various forms of muscular dystrophy [Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), sarcoglycanopathy, dysferlinopathy, calpainopathy, and merosin and collagen VI deficient congenital muscular dystrophies (CMDs)] and correlation of results with different histopathological features was investigated.

Results

Despite the fact that these diseases are not caused by defects of glycosyltransferases, decreased expression of LARGE was detected in many patient samples, partly correlating with the type of muscular dystrophy. Although immunolabelling of fully glycosylated αDG with VIA4–1 was reduced in dystrophinopathy patients, no significant relationship between reduction of LARGE expression and αDG hypoglycosylation was detected. Also, Merosin deficient CMD patients showed normal immunostaining with αDG despite severe reduction of LARGE expression.

Conclusions

Our data shows that it is not always possible to correlate LARGE expression and αDG glycosylation in different types of muscular dystrophies and suggests that there might be differences in αDG processing by LARGE which could be regulated under different pathological conditions.
Abkürzungen
ACTB
Beta-actin
BMD
Becker muscular dystrophy
CMD
Congenital muscular dystrophy
DAG1
DG-encoding gene
DG
Dystroglycan
DGC
Dystrophin-glycoprotein complex
DMD
Duchenne muscular dystrophy
ECM
Extracellular matrix
FCMD
Fukuyama congenital muscular dystrophy
FKRP
Fukutin-related protein
FKTN
Fukutin
H&E
Haematoxylin and eosin
LARGE
Like-acetylglucosaminyltransferase
LARGE-myd
myodystrophy mouse
LGMD
Limb-girdle muscular dystrophy
MEB
Muscle-eye-brain disease
POMGNT1
Protein-O-mannose ß2-N-acetylglucosaminyltransferase 1
POMT1
Protein-O-mannosyl transferase 1
POMT2
Protein-O-mannosyl transferase 2
qRT-PCR
Quantitative real-time PCR
WWS
Walker–Warburg syndrome
αDG
Alpha-dystroglycan

Background

Dystroglycan (DG), a heterodimeric transmembrane glycoprotein, is a central component of the dystrophin-glycoprotein complex (DGC) and responsible for a variety of physiological and developmental processes, including muscle stabilization, basement membrane assembly, cell migration and signalling [1]. The protein is synthesized as a precursor molecule that is post-translationally cleaved into a cell surface α- and a transmembrane β- subunits [2]. The extracellular subunit, alpha-dystroglycan (αDG), is a highly glycosylated basement membrane protein that acts as a receptor for non-collagenous proteins in the extracellular matrix (ECM) containing laminin globular domains, and for Old World arenaviruses [1, 35]. Mutations in the DG-encoding gene (DAG1) [6, 7] and several other genes whose products are involved, directly or indirectly, in glycosylation pathway of αDG [8] have been identified in various forms of congenital and limb-girdle muscular dystrophies (CMD/ LGMDs). These disorders are all associated with hypoglycosylation of αDG and a consequent lack of ligand-binding activity and thus are collectively termed as dystroglycanopathies [9].
Although the precise glycosylation pathway of αDG is not fully understood, the function of Like-acetylglucosaminyltransferase (LARGE) enzyme has been of particular interest. It is encoded by one of the largest genes (LARGE) in the human genome [1012], which is mutated in the myodystrophy mouse (LARGE-myd) [10, 13] and in patients affected by MDC1D, a subclass of CMD associated with skeletal muscle and structural brain involvement [11]. The mentioned enzyme is a bifunctional glycosyltransferase, with both xylosyltransferase and glucuronyltransferase activities and plays an important role in the glycosylation pathway of αDG. It transfers a novel heteropolysaccharide structure, repeating units of -glucuronic acid-β1,3-xylose-α1,3- to the basement membrane receptor DG [14]. This structure has been recently termed as matriglycan, which is bound to the αDG through a phosphorylated O-mannosyl glycan anchored at Thr317 and Thr319 in the mucin-like domain [8, 15, 16]. It is required for the αDG to bind laminin-G domain-containing ECM ligands including laminin, agrin, perlecan and neurexin [4, 1719]. It has been reported that LARGE-dependent matriglycan structure plays an important role for normal skeletal muscle function and the consequent reduction in the amount of glycans on αDG causes structural alterations of the basement membrane, immature neuromuscular junctions and dysfunctional muscle predisposed to dystrophy [20]. Uniquely, transient overexpression of LARGE has been shown to increase glycosylation of αDG as evaluated by increased immunoreactivity to antibodies IIH6 and VIA4–1 both of which are known to recognize carbohydrate moieties and leads to a recovery of receptor function in cells derived from patients diagnosed as Fukuyama CMD (FCMD), muscle-eye-brain disease (MEB) and Walker–Warburg syndrome (WWS) [17]. Also, the identical results has been reported in vivo, after adenovirus or adeno-associated virus mediated gene transfer of LARGE in Fukutin (FKTN), Fukutin-related protein (FKRP) and Protein-O-mannose ß2-N-acetylglucosaminyltransferase 1 (POMGNT1) deficient mice models [2123]. Although a recent study demonstrates that fukutin is required for the ability of LARGE to hyperglycosylate αDG [24], this strategy is already regarded as a promising therapeutic approach for preventing/slowing progression of a broad range of dystroglycanopathies regardless of the causative gene defects. However, it remains crucial to analyze such a strategy in different types of muscular dystrophies other than dystroglycanopathy.
In this study, we analyzed the influence of LARGE gene expression profile on VIA4–1 immunoreactivity in different types of muscular dystrophies, except for dystroglycanopathies [i.e. Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), sarcoglycanopathy, dysferlinopathy, calpainopathy, and merosin and collagen VI deficient CMDs]. Correlation between different histopathological features, LARGE expression profile and glycosylation status of αDG was investigated. We detected reduced expression level of LARGE in different types of muscular dystrophies, partly correlating with the severity of dystrophic changes, but we did not find any significant relationship between reduction of LARGE expression and VIA4–1 immunoreactivity.

Methods

Patients and muscle biopsies

The study protocol was approved by the Hacettepe University Faculty of Medicine Ethical Review Board (FON 10/35–11). As the legal age of consent is 18 years of age in Turkey, written informed consent was obtained from patients’ parents or legal guardians at the time of diagnostic muscle biopsy. Skeletal muscle biopsies from 3 controls and 26 patients with clinicopathological and/or molecular diagnosis of various forms of muscular dystrophy other than dystroglycanopathy were included in this study. The number of patients in each disease type is indicated in parentheses; DMD (3), BMD (4), sarcoglycanopathy (5), dysferlinopathy (1), calpainopathy (1), Merosin deficient CMD (4), Collagen VI deficient CMD (3), unclassified CMD (2), and unclassified MD (3). All patients have been investigated in the Department of Pediatrics at Hacettepe University Faculty of Medicine and open biopsies were taken from vastus lateralis muscle at the time of diagnosis. The muscle biopsy specimens were rapidly frozen in isopentane cooled in liquid nitrogen and kept at − 80 °C until use. Serial transverse muscle sections were cut by cryostat for histochemical staining. Standard histological and histochemical techniques were applied to cryostat sections, including Haematoxylin and eosin (H&E), modified Gomori trichrome, periodic acid Schiff, oil-red-O, nicotinamide adenine dinucleotide dehydrogenase-tetrazolium reductase, succinic dehydrogenase, cytochrome-c oxidase and ATPase. Immunohistochemical and/or immunofluorescent studies were done at the time of diagnosis, using antibodies against dystrophins, sarcoglycans, dysferlin, laminin α2 (merosin), collagen VI, and αDG, as appropriate.

Histopathological evaluation

H&E stained archive sections were examined to identify the severity of various dystrophic changes, including necrosis, regeneration, adiposis, endomysial and perimysial fibrosis and inflammation. These pathological changes were scored from 0 (absent) to 4 (severe).

Immunofluorescent staining

For immunofluorescent staining, skeletal muscle cryosections of 7 μm were immunostained with primary antibodies [1/10 for mouse anti-α-Dystroglycan antibody (clone VIA4–1 against fully glycosylated form of αDG, Millipore), 1/50 for sheep anti-α-Dystroglycan core antibody (R&D Systems: AF6868), 1/100 for mouse anti-Spectrin (NCL-SPEC1 Clone: RBC2/3D5, Novocastra), 1:20 for anti-Beta-Dystroglycan (βDG) (NCL-b-DG Clone 43DAG1/8D5, Novocastra) and 1:1000 for mouse anti-laminin α2 (MAB1922 Chemicon (Millipore)] for 1 h at room temperature, followed by incubation with Alexa Fluor 488 goat anti-mouse or donkey anti-sheep IgG antibodies (Molecular Probes) at 1:1000 dilution for 45 min at room temperature. All dilutions and washing steps were made in phosphate buffered saline. Immunostained sections were then mounted and observed with epifluorescence using a Nicon Eclipse E400 microscope. The fluorescent labeling was scored from 0 (absent) to 3 (intense) using a scale based on the proportion of positive fibers and intensity relative to the control muscle sections.

Quantitative real-time PCR (qRT-PCR)

In order to assess the expression of LARGE, total RNA was extracted from liquid nitrogen fresh-frozen skeletal muscle tissues (3 mm3) from 26 patiens and 3 control samples using RNeasy® fibrous tissue mini kit (Qiagen) and cDNA was synthesized from 500 ng RNA using QuantiTect Reverse Transcription kit (Qiagen) according to manufacturer’s guidelines. Real-time PCR was then performed using SYBR Green JumpStart TaqReadyMix kit (Sigma) on Rotor-Gene 6000™ (Corbett Research) with the following conditions: 2 min at 94 °C, followed by 40 cycles of 5 s at 94 °C, 10 s at 58 °C, and 15 s at 72 °C. Reactions were performed in a total volume of 10 μL, including 5 μL SYBR Green JumpStart TaqReadyMix kit (Sigma), 1.2 μl of 25 mM MgCl2 (Sigma), 0.4 μl each of 10 mM forward and reverse primers and 1 μl of template cDNA. Mastermix with no cDNA was used as the no template control whereas water was used as the negative control. The relative amount of mRNA, normalized to an internal control human beta-actin (ACTB) and relative to a calibrator (control), was calculated by 2-∆∆CT. Primers used were as follows: LARGE-F: 5’-TGAGCCGTATGTTGTTGTGAGAC-3′; LARGE-R: 5’-GATGCGGTATTGCTTGTTGGAAC-3′ and ACTB-F: 5′- CGCAAAGACCTGTACGCCAAC -3′; ACTB-R: 5′- GAGCCGCCGATCCACACG -3′. All reactions were performed in triplicate and optimal threshold in 95–100% efficiency. All data was analyzed using Rotor-Gene 6000 Series Software 1.7 (Corbett Research).

Statistical analysis

All experiments performed at least three times and data were evaluated as means ± SD. The significance of differences between experimental groups was determined using Mann-Whitney U test. p values less than 0.05 were considered statistically significant.

Results

We assessed human muscle biopsies from DMD, BMD, sarcoglycanopathy, calpainopathy, dysferlinopathy, and merosin and collagen VI deficient CMDs, in addition to unclassified CMDs and MDs. Scoring for the severity of histopathological features and age at biopsy for each patient are shown in Table 1. In each muscular dystrophy group, samples had variable severity of histopathological findings such as necrosis, regeneration, fibrosis, adiposis and inflammation. Even some patients with the same diagnosis and similar age of biopsy showed different severity of pathological changes.
Table 1
The clinicopathological diagnosis and histopathological characteristics of the patients (Assessment scores: 0: absent; 1: rare; 2: mild; 3: moderate; 4: severe)
https://static-content.springer.com/image/art%3A10.1186%2Fs12883-018-1207-0/MediaObjects/12883_2018_1207_Tab1_HTML.png
We first assessed the expression level of LARGE by qRT-PCR and detected reduced expression level of LARGE gene in analyzed samples, partly correlating with the form of muscular dystrophy (Fig. 1). The reduction of LARGE expression was more significant in all cases of DMD (P1, P2 and P3) and merosin deficient CMD (P14, P15, P16 and P17) as compared to control skeletal muscle. Also, LARGE mRNA levels were markedly reduced in all BMD cases, except one (P6). The expression of LARGE was similarly reduced in two cases of unclassified CMD (P22 and P23) and MD (P24 and P25), who had similar histopathological patterns. However, decreased expression in one case of Collagen VI deficient CMD (P18) with mild dystrophic features remains unexplained. The results of our study revealed different expression profiles for LARGE also among patients with different types of sarcoglycanopathies. Reduced LARGE expression levels were found also in the single dysferlinopathy (P13) and calpainopathy (P21) patients compared with age-matched healthy controls, with more severe reduction in LARGE and more severe dystrophic features in the case with dysferlin deficiency.
In order to reveal a correlation between LARGE expression and functional glycosylation of αDG, skeletal muscle sections were immunostained using VIA4–1 antibody against fully glycosylated form of αDG, and evaluated together with αDG core protein (Fig. 2), beta-dystroglycan and laminin α2 immunostaining. The spectrum of VIA4–1 labeling was very variable and ranged from an absent to intense, compared with control skeletal muscle, whereas the immunoreactivity of core protein was indistinguishable between control and patients (Fig. 3). Therefore, the loss of VIA4–1 reactivity in some specimens reflects a reduction of α-DG glycosylation, rather than loss of α-DG core protein expression. The skeletal muscle of patients with DMD showed a marked depletion of αDG glycosylation. The reduction of VIA4–1 labeling in BMD group ranged from an absence on most fibres to a very mild reduction. Also, in some patients with sarcoglycanopathy, Collagen VI deficient CMD and unclassified MD, a mild reduction of sarcolemmal labelling with VIA4–1 antibody was detected. Interestingly, patient 8 and 18 displayed an almost total absence of αDG glycosylation relative to other patients within their group. Spectrin appeared normal in all cases indicating that the reduction in αDG hypoglycosylation was not due to non-specific damage of the plasma membrane. Except for merosin deficient CMDs, laminin α2 staining was also normal in all analyzed cases. Labeling of the transmembrane protein βDG was preserved in most cases, although a mild reduction has been reported in 9 of the 26 cases, mostly in dystrophinopathies. There was no significant relationship between reduction of LARGE expression and αDG hypoglycosylation, most obvious example being the merosin deficient CMD patients (P14, P15, P16 and P17) (Figs. 1 and 3) who showed normal immunostaining with αDG despite severe reduction of LARGE expression.

Discussion

In order to determine correlations between LARGE expression, glycosylation of αDG and histopathological parameters, we analyzed LARGE gene expression profile and glycosylation status of αDG in various forms of muscular dystrophies, except for dystroglycanopathies. The expression level of LARGE was analyzed by qRT-PCR in skeletal muscle biopsies from 26 patients diagnosed as different forms of muscular dystrophy with various histopathological phenotypes (Table 1). Our findings indicated that most of the patients with severe histopathological phenotype (DMD and merosin deficient CMD) have decreased LARGE expression levels compared to control skeletal muscle (Fig. 1). We then evaluated whether there was a relationship between LARGE expression and αDG glycosylation. The functional glycosylation of αDG is associated with the binding of either IIH6 and/or VIA4–1 antibodies to αDG. Thus, immunofluorescence staining was done using the widely used VIA4–1 antibody. Our findings indicate that irrespective of the muscular dystrophy type, the degree of hypoglycosylation varied (Figs. 2 and 3) and there was generally no correlation with clinical/histopathological severity. Similar results have previously been reported in dystroglycanopathy patients with Protein-O-mannosyl transferase 1 (POMT1), Protein-O-mannosyl transferase 2 (POMT2), POMGnT1, FKTN and FKRP mutations [25]. Also, our results were not consistent with the findings which suggests a correlation between a reduction in αDG labeling and the clinical severity in mild, late-onset LGMD type 2I and severe CMD [20, 26]. On the other hand, we observed a severe reduction of αDG glycosylation in skeletal muscle biopsies of dystrophinopathy patients as a secondary finding, which has already been shown in DMD patients and mdx mouse [27, 28]. In contrast to those findings, the labelling obtained using the αDG core antibody was preserved in all patients, which suggested that the glycosylation of αDG is affected independently of DG protein expression.
In dystroglycanopathies, the effects of decreased glycosylation on laminin binding are variable [4, 11]. Therefore, further studies are necessary to evaluate if hypoglycosylation of αDG affects laminin binding or other functions of αDG in different types of muscular dystrophies other than dystroglycanopathy. Campbell and colleagues have previously demonstrated that short LARGE-glycan repeats reduces normal physiological function of muscle and predisposes it to dystrophy and showed that upregulation of LARGE and DG facilitates extension of LARGE-glycan repeat chains in differentiating mouse muscle [20]. However, consistent with our preliminary findings [29], we did not find any significant relationship between reduction of LARGE expression and αDG hypoglycosylation (Figs. 1 and 3), except for DMD. Our results suggest that LARGE enzyme may have an additional function in skeletal muscle fibers that is probably distinct from adding a critical sugar chain onto αDG. Equally likely is that the RNA changes in LARGE, while present, are not related at all to the MD phenotype. Transcriptional changes are difficult to interpret in prediction effects downstream.

Conclusions

Glycosylation of αDG undergoes tissue and developmental stage specific changes, which indicates that it may have different roles in developing and fully differentiated skeletal muscle [4, 30]. αDG glycosylation is likely to be regulated by developmental and differential expression of numerous enzymes, including LARGE. Therefore the detected LARGE expression profiles in various form of muscular dystrophies may still reflect responses to pathological processes of skeletal muscle related to αDG glycosylation. Our cohort encompassed a wide range of muscular dystrophies, suggesting that a different pathomechanism may explain the relationship between LARGE expression and αDG glycosylation in the pathogenesis of different types of muscular dystrophies and highlighting the need for further studies.

Acknowledgements

Not applicable.

Funding

This study was supported by a grant from Hacettepe University Scientific Research Projects Coordination Unit (grant number 011 D01 101 003).

Availability of data and materials

The datasets analyzed and/or generated during the current study are available from the corresponding author on reasonable request.
This study was performed with the approval of the Ethical Review Board of Hacettepe University Faculty of Medicine, Ankara, Turkey (FON 10/35–11). As the legal age of consent is 18 years of age in Turkey, written informed consent was obtained from patients’ parents or legal guardians at the time of diagnostic muscle biopsy. All studies were conducted in accordance with the principles of the Declaration of Helsinki.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Barresi R, Campbell KP. Dystroglycan: from biosynthesis to pathogenesis of human disease. J Cell Sci. 2006;119:199–207.CrossRef Barresi R, Campbell KP. Dystroglycan: from biosynthesis to pathogenesis of human disease. J Cell Sci. 2006;119:199–207.CrossRef
2.
Zurück zum Zitat Ibraghimov-Beskrovnaya O, Ervasti JM, Leveille CJ, Slaughter CA, Sernett SW, Campbell KP. Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix. Nature. 1992;355:696–702.CrossRef Ibraghimov-Beskrovnaya O, Ervasti JM, Leveille CJ, Slaughter CA, Sernett SW, Campbell KP. Primary structure of dystrophin-associated glycoproteins linking dystrophin to the extracellular matrix. Nature. 1992;355:696–702.CrossRef
3.
Zurück zum Zitat Cao W, Henry MD, Borrow P, Yamada H, Elder JH, Ravkov EV, Nichol ST, Compans RW, Campbell KP, Oldstone MB. Identification of α-dystroglycan as a receptor for lymphocytic choriomeningitis virus and Lassa fever virus. Science. 1998;282:2079–81.CrossRef Cao W, Henry MD, Borrow P, Yamada H, Elder JH, Ravkov EV, Nichol ST, Compans RW, Campbell KP, Oldstone MB. Identification of α-dystroglycan as a receptor for lymphocytic choriomeningitis virus and Lassa fever virus. Science. 1998;282:2079–81.CrossRef
4.
Zurück zum Zitat Michele DE, Barresi R, Kanagawa M, Saito F, Cohn RD, Satz JS, Dollar J, Nishino I, Kelley RI, Somer H, Straub V, Mathews KD, Moore SA, Campbell KP. Post-translational disruption of dystroglycan-ligand interactions in congenital muscular dystrophies. Nature. 2002;418:417–22.CrossRef Michele DE, Barresi R, Kanagawa M, Saito F, Cohn RD, Satz JS, Dollar J, Nishino I, Kelley RI, Somer H, Straub V, Mathews KD, Moore SA, Campbell KP. Post-translational disruption of dystroglycan-ligand interactions in congenital muscular dystrophies. Nature. 2002;418:417–22.CrossRef
5.
Zurück zum Zitat Spiropoulou CF, Kunz S, Rollin PE, Campbell KP, Oldstone MB. New World arenavirus clade C, but not clade A and B viruses, utilizes alpha-dystroglycan as its major receptor. J Virol. 2002;76:5140–6.CrossRef Spiropoulou CF, Kunz S, Rollin PE, Campbell KP, Oldstone MB. New World arenavirus clade C, but not clade A and B viruses, utilizes alpha-dystroglycan as its major receptor. J Virol. 2002;76:5140–6.CrossRef
6.
Zurück zum Zitat Dinçer P, Balci B, Yuva Y, Talim B, Brockington M, Dinçel D, Torelli S, Brown S, Kale G, Haliloglu G, Gerçeker FO, Atalay RC, Yakicier C, Longman C, Muntoni F, Topaloglu H. A novel form of recessive limb girdle muscular dystrophy with mental retardation and abnormal expression of alpha-dystroglycan. Neuromuscul Disord. 2003;13:771–8.CrossRef Dinçer P, Balci B, Yuva Y, Talim B, Brockington M, Dinçel D, Torelli S, Brown S, Kale G, Haliloglu G, Gerçeker FO, Atalay RC, Yakicier C, Longman C, Muntoni F, Topaloglu H. A novel form of recessive limb girdle muscular dystrophy with mental retardation and abnormal expression of alpha-dystroglycan. Neuromuscul Disord. 2003;13:771–8.CrossRef
7.
Zurück zum Zitat Hara Y, Balci-Hayta B, Yoshida-Moriguchi T, Kanagawa M, Beltrán-Valero de Bernabé D, Gündeşli H, Willer T, Satz JS, Crawford RW, Burden SJ, Kunz S, Oldstone MB, Accardi A, Talim B, Muntoni F, Topaloğlu H, Dinçer P, Campbell KP. A dystroglycan mutation associated with limb-girdle muscular dystrophy. N Engl J Med. 2011;364:939–46.CrossRef Hara Y, Balci-Hayta B, Yoshida-Moriguchi T, Kanagawa M, Beltrán-Valero de Bernabé D, Gündeşli H, Willer T, Satz JS, Crawford RW, Burden SJ, Kunz S, Oldstone MB, Accardi A, Talim B, Muntoni F, Topaloğlu H, Dinçer P, Campbell KP. A dystroglycan mutation associated with limb-girdle muscular dystrophy. N Engl J Med. 2011;364:939–46.CrossRef
8.
Zurück zum Zitat Yoshida-Moriguchi T, Campbell KP. Matriglycan: a novel polysaccharide that links dystroglycan to the basement membrane. Glycobiology. 2015;25:702–13.CrossRef Yoshida-Moriguchi T, Campbell KP. Matriglycan: a novel polysaccharide that links dystroglycan to the basement membrane. Glycobiology. 2015;25:702–13.CrossRef
9.
Zurück zum Zitat Godfrey C, Foley AR, Clement E, Muntoni F. Dystroglycanopathies: coming into focus. Curr Opin Genet Dev. 2011;21:278–85.CrossRef Godfrey C, Foley AR, Clement E, Muntoni F. Dystroglycanopathies: coming into focus. Curr Opin Genet Dev. 2011;21:278–85.CrossRef
10.
Zurück zum Zitat Grewal PK, Hewitt JE. Mutation of Large, which encodes a putative glycosyltransferase, in an animal model of muscular dystrophy. Biochim Biophys Acta. 2002;1573:216–24.CrossRef Grewal PK, Hewitt JE. Mutation of Large, which encodes a putative glycosyltransferase, in an animal model of muscular dystrophy. Biochim Biophys Acta. 2002;1573:216–24.CrossRef
11.
Zurück zum Zitat Longman C, Brockington M, Torelli S, Jimenez-Mallebrera C, Kennedy C, Khalil N, Feng L, Saran RK, Voit T, Merlini L, Sewry CA, Brown SC, Muntoni F. Mutations in the human LARGE gene cause MDC1D, a novel form of congenital muscular dystrophy with severe mental retardation and abnormal glycosylation of alpha-dystroglycan. Hum Mol Genet. 2003;12:2853–61.CrossRef Longman C, Brockington M, Torelli S, Jimenez-Mallebrera C, Kennedy C, Khalil N, Feng L, Saran RK, Voit T, Merlini L, Sewry CA, Brown SC, Muntoni F. Mutations in the human LARGE gene cause MDC1D, a novel form of congenital muscular dystrophy with severe mental retardation and abnormal glycosylation of alpha-dystroglycan. Hum Mol Genet. 2003;12:2853–61.CrossRef
12.
Zurück zum Zitat van Reeuwijk J, Grewal PK, Salih MA, Beltrán-Valero de Bernabé D, JM ML, Michielse CB, Herrmann R, Hewitt JE, Steinbrecher A, Seidahmed MZ, Shaheed MM, Abomelha A, Brunner HG, van Bokhoven H, Voit T. Intragenic deletion in the LARGE gene causes Walker-Warburg syndrome. Hum Genet. 2007;121:685–90.CrossRef van Reeuwijk J, Grewal PK, Salih MA, Beltrán-Valero de Bernabé D, JM ML, Michielse CB, Herrmann R, Hewitt JE, Steinbrecher A, Seidahmed MZ, Shaheed MM, Abomelha A, Brunner HG, van Bokhoven H, Voit T. Intragenic deletion in the LARGE gene causes Walker-Warburg syndrome. Hum Genet. 2007;121:685–90.CrossRef
13.
Zurück zum Zitat Grewal PK, Holzfeind PJ, Bittner RE, Hewitt JE. Mutant glycosyltransferase and altered glycosylation of alpha-dystroglycan in the myodystrophy mouse. Nat Genet. 2001;28:151–4.CrossRef Grewal PK, Holzfeind PJ, Bittner RE, Hewitt JE. Mutant glycosyltransferase and altered glycosylation of alpha-dystroglycan in the myodystrophy mouse. Nat Genet. 2001;28:151–4.CrossRef
14.
Zurück zum Zitat Inamori K, Yoshida-Moriguchi T, Hara Y, Anderson ME, Yu L, Campbell KP. Dystroglycan function requires xylosyl- and glucuronyltransferase activities of LARGE. Science. 2012;335:93–6.CrossRef Inamori K, Yoshida-Moriguchi T, Hara Y, Anderson ME, Yu L, Campbell KP. Dystroglycan function requires xylosyl- and glucuronyltransferase activities of LARGE. Science. 2012;335:93–6.CrossRef
15.
Zurück zum Zitat Hara Y, Kanagawa M, Kunz S, Yoshida-Moriguchi T, Satz JS, Kobayashi YM, Zhu Z, Burden SJ, Oldstone MB, Campbell KP. Like-acetylglucosaminyltransferase (LARGE)-dependent modification of dystroglycan at Thr-317/319 is required for laminin binding and arenavirus infection. Proc Natl Acad Sci U S A. 2011;108:17426–31.CrossRef Hara Y, Kanagawa M, Kunz S, Yoshida-Moriguchi T, Satz JS, Kobayashi YM, Zhu Z, Burden SJ, Oldstone MB, Campbell KP. Like-acetylglucosaminyltransferase (LARGE)-dependent modification of dystroglycan at Thr-317/319 is required for laminin binding and arenavirus infection. Proc Natl Acad Sci U S A. 2011;108:17426–31.CrossRef
16.
Zurück zum Zitat Yoshida-Moriguchi T, Yu L, Stalnaker SH, Davis S, Kunz S, Madson M, Oldstone MB, Schachter H, Wells L, Campbell KP. O-mannosyl phosphorylation of alpha-dystroglycan is required for laminin binding. Science. 2010;327:88–92.CrossRef Yoshida-Moriguchi T, Yu L, Stalnaker SH, Davis S, Kunz S, Madson M, Oldstone MB, Schachter H, Wells L, Campbell KP. O-mannosyl phosphorylation of alpha-dystroglycan is required for laminin binding. Science. 2010;327:88–92.CrossRef
17.
Zurück zum Zitat Barresi R, Michele DE, Kanagawa M, Harper HA, Dovico SA, Satz JS, Moore SA, Zhang W, Schachter H, Dumanski JP, Cohn RD, Nishino I, Campbell KP. LARGE can functionally bypass α-dystroglycan glycosylation defects in distinct congenital muscular dystrophies. Nature Med. 2004;10:696–703.CrossRef Barresi R, Michele DE, Kanagawa M, Harper HA, Dovico SA, Satz JS, Moore SA, Zhang W, Schachter H, Dumanski JP, Cohn RD, Nishino I, Campbell KP. LARGE can functionally bypass α-dystroglycan glycosylation defects in distinct congenital muscular dystrophies. Nature Med. 2004;10:696–703.CrossRef
18.
Zurück zum Zitat Sato S, Omori Y, Katoh K, Kondo M, Kanagawa M, Miyata K, Funabiki K, Koyasu T, Kajimura N, Miyoshi T, Sawai H, Kobayashi K, Tani A, Toda T, Usukura J, Tano Y, Fujikado T, Furukawa T. Pikachurin, a dystroglycan ligand, is essential for photoreceptor ribbon synapse formation. Nature Neurosci. 2008;11:923–31.CrossRef Sato S, Omori Y, Katoh K, Kondo M, Kanagawa M, Miyata K, Funabiki K, Koyasu T, Kajimura N, Miyoshi T, Sawai H, Kobayashi K, Tani A, Toda T, Usukura J, Tano Y, Fujikado T, Furukawa T. Pikachurin, a dystroglycan ligand, is essential for photoreceptor ribbon synapse formation. Nature Neurosci. 2008;11:923–31.CrossRef
19.
Zurück zum Zitat Willer T, Valero MC, Tanner W, Cruces J, Strahl S. O-mannosyl glycans: from yeast to novel associations with human disease. Cur Opin Struct Biol. 2003;13:621–30.CrossRef Willer T, Valero MC, Tanner W, Cruces J, Strahl S. O-mannosyl glycans: from yeast to novel associations with human disease. Cur Opin Struct Biol. 2003;13:621–30.CrossRef
20.
Zurück zum Zitat Goddeeris MM, Wu B, Venzke D, Yoshida-Moriguchi T, Saito F, Matsumura K, Moore SA, Campbell KP. LARGE glycans on dystroglycan function as a tunable matrix scaffold to prevent dystrophy. Nature. 2013;503:136–40.CrossRef Goddeeris MM, Wu B, Venzke D, Yoshida-Moriguchi T, Saito F, Matsumura K, Moore SA, Campbell KP. LARGE glycans on dystroglycan function as a tunable matrix scaffold to prevent dystrophy. Nature. 2013;503:136–40.CrossRef
21.
Zurück zum Zitat Kanagawa M, Nishimoto A, Chiyonobu T, Takeda S, Miyagoe-Suzuki Y, Wang F, Fujikake N, Taniguchi M, Lu Z, Tachikawa M, Nagai Y, Tashiro F, Miyazaki J, Tajima Y, Takeda S, Endo T, Kobayashi K, Campbell KP, Toda T. Residual laminin-binding activity and enhanced dystroglycan glycosylation by LARGE in novel model mice to dystroglycanopathy. Hum Mol Genet. 2009;18:621–31.CrossRef Kanagawa M, Nishimoto A, Chiyonobu T, Takeda S, Miyagoe-Suzuki Y, Wang F, Fujikake N, Taniguchi M, Lu Z, Tachikawa M, Nagai Y, Tashiro F, Miyazaki J, Tajima Y, Takeda S, Endo T, Kobayashi K, Campbell KP, Toda T. Residual laminin-binding activity and enhanced dystroglycan glycosylation by LARGE in novel model mice to dystroglycanopathy. Hum Mol Genet. 2009;18:621–31.CrossRef
22.
Zurück zum Zitat Vannoy CH, Xu L, Keramaris E, Lu P, Xiao X, Lu QL. Adeno-associated virus-mediated overexpression of LARGE rescues α-dystroglycan function in dystrophic mice with mutations in the fukutin-related protein. Hum Gene Ther Method. 2014;25:187–96.CrossRef Vannoy CH, Xu L, Keramaris E, Lu P, Xiao X, Lu QL. Adeno-associated virus-mediated overexpression of LARGE rescues α-dystroglycan function in dystrophic mice with mutations in the fukutin-related protein. Hum Gene Ther Method. 2014;25:187–96.CrossRef
23.
Zurück zum Zitat Yu M, He Y, Wang K, Zhang P, Zhang S, Hu H. Adeno-associated viral-mediated LARGE gene therapy rescues the muscular dystrophic phenotype in mouse models of dystroglycanopathy. Hum Gene Ther. 2013;24:317–30.CrossRef Yu M, He Y, Wang K, Zhang P, Zhang S, Hu H. Adeno-associated viral-mediated LARGE gene therapy rescues the muscular dystrophic phenotype in mouse models of dystroglycanopathy. Hum Gene Ther. 2013;24:317–30.CrossRef
24.
Zurück zum Zitat Ohtsuka Y, Kanagawa M, Yu CC, Ito C, Chiyo T, Kobayashi K, Okada T, Takeda S, Toda T. Fukutin is prerequisite to ameliorate muscular dystrophic phenotype by myofiber-selective LARGE expression. Sci Rep. 2015;5:8316.CrossRef Ohtsuka Y, Kanagawa M, Yu CC, Ito C, Chiyo T, Kobayashi K, Okada T, Takeda S, Toda T. Fukutin is prerequisite to ameliorate muscular dystrophic phenotype by myofiber-selective LARGE expression. Sci Rep. 2015;5:8316.CrossRef
25.
Zurück zum Zitat Jimenez-Mallebrera C, Torelli S, Feng L, Kim J, Godfrey C, Clement E, Mein R, Abbs S, Brown SC, Campbell KP, Kröger S, Talim B, Topaloglu H, Quinlivan R, Roper H, Childs AM, Kinali M, Sewry CA, Muntoni F. A comparative study of alpha-dystroglycan glycosylation in dystroglycanopathies suggests that the hypoglycosylation of alpha-dystroglycan does not consistently correlate with clinical severity. Brain Pathol. 2009;19:596–611.CrossRef Jimenez-Mallebrera C, Torelli S, Feng L, Kim J, Godfrey C, Clement E, Mein R, Abbs S, Brown SC, Campbell KP, Kröger S, Talim B, Topaloglu H, Quinlivan R, Roper H, Childs AM, Kinali M, Sewry CA, Muntoni F. A comparative study of alpha-dystroglycan glycosylation in dystroglycanopathies suggests that the hypoglycosylation of alpha-dystroglycan does not consistently correlate with clinical severity. Brain Pathol. 2009;19:596–611.CrossRef
26.
Zurück zum Zitat Brown SC, Torelli S, Brockington M, Yuva Y, Jimenez C, Feng L, Anderson L, Ugo I, Kroger S, Bushby K, Voit T, Sewry C, Muntoni F. Abnormalities in alpha-dystroglycan expression in MDC1C and LGMD2I muscular dystrophies. Am J Pathol. 2004;164:727–37.CrossRef Brown SC, Torelli S, Brockington M, Yuva Y, Jimenez C, Feng L, Anderson L, Ugo I, Kroger S, Bushby K, Voit T, Sewry C, Muntoni F. Abnormalities in alpha-dystroglycan expression in MDC1C and LGMD2I muscular dystrophies. Am J Pathol. 2004;164:727–37.CrossRef
27.
Zurück zum Zitat Ohlendieck K, Campbell KP. Dystrophin-associated proteins are greatly reduced in skeletal muscle from mdx mice. J Cell Biol. 1991;115:1685–94.CrossRef Ohlendieck K, Campbell KP. Dystrophin-associated proteins are greatly reduced in skeletal muscle from mdx mice. J Cell Biol. 1991;115:1685–94.CrossRef
28.
Zurück zum Zitat Pereira CC, Kiyomoto BH, Cardoso R, Oliveira AS. Duchenne muscular dystrophy: alpha-dystroglycan immunoexpression in skeletal muscle and cognitive performance. Arq Neuropsiquiatr. 2005;63:984–9.CrossRef Pereira CC, Kiyomoto BH, Cardoso R, Oliveira AS. Duchenne muscular dystrophy: alpha-dystroglycan immunoexpression in skeletal muscle and cognitive performance. Arq Neuropsiquiatr. 2005;63:984–9.CrossRef
29.
Zurück zum Zitat Balci-Hayta B, Talim B, Topaloglu H, Kale G, Dincer P. Reduction of LARGE expression in different types of muscular dystrophies other than dystroglycanopathy. Neuromuscul Disord. 2013;23(9–10):p780.CrossRef Balci-Hayta B, Talim B, Topaloglu H, Kale G, Dincer P. Reduction of LARGE expression in different types of muscular dystrophies other than dystroglycanopathy. Neuromuscul Disord. 2013;23(9–10):p780.CrossRef
30.
Zurück zum Zitat Ervasti JM, Burwell AL, Geissler AL. Tissue-specific heterogeneity in alphadystroglycan sialoglycosylation. Skeletal muscle alpha-dystroglycan is a latent receptor for Vicia villosa agglutinin b4 masked by sialic acid modification. J Biol Chem. 1997;272:22315–21.CrossRef Ervasti JM, Burwell AL, Geissler AL. Tissue-specific heterogeneity in alphadystroglycan sialoglycosylation. Skeletal muscle alpha-dystroglycan is a latent receptor for Vicia villosa agglutinin b4 masked by sialic acid modification. J Biol Chem. 1997;272:22315–21.CrossRef
Metadaten
Titel
LARGE expression in different types of muscular dystrophies other than dystroglycanopathy
verfasst von
Burcu Balci-Hayta
Beril Talim
Gulsev Kale
Pervin Dincer
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
BMC Neurology / Ausgabe 1/2018
Elektronische ISSN: 1471-2377
DOI
https://doi.org/10.1186/s12883-018-1207-0

Weitere Artikel der Ausgabe 1/2018

BMC Neurology 1/2018 Zur Ausgabe