functional requirements for fukutin-related protein in the golgi

13
Functional requirements for fukutin-related protein in the Golgi apparatus Chris T. Esapa 1,{ , Matthew A. Benson 1,{ , Jo ¨ rn E. Schro ¨ der 2 , Enca Martin-Rendon 1,{ , Martin Brockington 3 , Susan C. Brown 3 , Francesco Muntoni 3 , Stephan Kro ¨ ger 2 and Derek J. Blake 1, * 1 Department of Pharmacology, University of Oxford, Oxford, UK, 2 Institute for Physiological Chemistry, University of Mainz, Mainz, Germany and 3 The Dubowitz Neuromuscular Centre, Department of Paediatrics, Imperial College School of Medicine, Hammersmith Hospital Campus, London, UK Received August 22, 2002; Revised and Accepted October 21, 2002 Two forms of congenital muscular dystrophy (CMD), Fukuyama CMD and CMD type 1C (MDC1C) are caused by mutations in the genes encoding two putative glycosyltransferases, fukutin and fukutin-related protein (FKRP). Additionally, mutations in the FKRP gene also cause limb-girdle muscular dystrophy type 2I (LGMD2I), a considerably milder allelic variant than MDC1C. All of these diseases are associated with secondary changes in muscle a-dystroglycan expression. To elucidate the function of FKRP and fukutin and examine the effects of MDC1C patient mutations, we have determined the mechanism for the subcellular location of each protein. FKRP and fukutin are targeted to the medial-Golgi apparatus through their N-termini and transmembrane domains. Overexpression of FKRP in CHO cells alters the post-translational processing of a- and b-dystroglycan inhibiting maturation of the two isoforms. Mutations in the DxD motif in the putative active site of the protein or in the Golgi-targeting sequence, which cause FKRP to be inefficiently trafficked to the Golgi apparatus, did not alter dystroglycan processing in vitro. The P448L mutation in FKRP that causes congenital muscular dystrophy changes a conserved amino acid resulting in the mislocalization of the mutant protein in the cell that is unable to alter dystroglycan processing. Our data show that FKRP and fukutin are Golgi-resident proteins and that FKRP is required for the post-translational modification of dystroglycan. Aberrant processing of dystroglycan caused by a mislocalized FKRP mutant could be a novel mechanism that causes congenital muscular dystrophy. INTRODUCTION The congenital muscular dystrophies (CMDs) are a hetero- geneous group of disorders that are characterized by early onset muscular dystrophy (reviewed in 1,2). Several of these disorders are also associated with brain defects, mental retardation and abnormalities of the visual system (1–3). Recently, the genes for some of these diseases have been iden- tified (4–6). One of the most severe of these disorders is Fukuyama congenital muscular dystrophy (FCMD), which is caused by mutations in the gene encoding fukutin. FCMD is the second most common muscular dystrophy in the Japanese population and predominates in populations originating from North East Asia (7). FCMD patients are more severely affected than those with Duchenne muscular dystrophy (DMD) and often never walk. These patients are mentally retarded, affected by epilepsy and have severe morphological brain abnormalities indicative of a defect in cortical neuronal migration (8). The majority of FCMD patients have inherited the same mutation, namely the insertion of a retrotransposon in the 3 0 untranslated region of the FCMD gene (4). Compound heterozygotes have also been described that have point mutations in one allele of the FCMD gene and the retrotransposon on the second allele (9). However, no patients have been described that are homozygous for the point mutation suggesting that the complete lack of fukutin could result in embryonic lethality (9). The FCMD gene was predicted to encode a secreted protein called fukutin. However, due to the lack of suitable antibodies, the subcellular location of fukutin has not been determined (4). Interestingly, muscle biopsies from FCMD patients show a dramatic reduction in glycosylated a-dystroglycan, a com- ponent of the dystrophin protein complex (see 10 for a *To whom correspondence should be addressed at: Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK. Tel: þ44 01865271860; Fax: þ44 01865271853; Email: [email protected] { Present address: National Blood Service, John Radcliffe Hospital, Oxford, OX3 9DU, UK. { The authors wish it to be known that, in their opinion, these two authors should be considered as joint First Authors. # 2002 Oxford University Press Human Molecular Genetics, 2002, Vol. 11, No. 26 3319–3331 Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489 by guest on 11 February 2018

Upload: nguyendiep

Post on 01-Jan-2017

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Functional requirements for fukutin-related protein in the Golgi

Functional requirements for fukutin-relatedprotein in the Golgi apparatus

Chris T. Esapa1,{, Matthew A. Benson1,{, Jorn E. Schroder2, Enca Martin-Rendon1,{,

Martin Brockington3, Susan C. Brown3, Francesco Muntoni3, Stephan Kroger2 and

Derek J. Blake1,*

1Department of Pharmacology, University of Oxford, Oxford, UK, 2Institute for Physiological Chemistry,

University of Mainz, Mainz, Germany and 3The Dubowitz Neuromuscular Centre, Department of Paediatrics,

Imperial College School of Medicine, Hammersmith Hospital Campus, London, UK

Received August 22, 2002; Revised and Accepted October 21, 2002

Two forms of congenital muscular dystrophy (CMD), Fukuyama CMD and CMD type 1C (MDC1C) are causedby mutations in the genes encoding two putative glycosyltransferases, fukutin and fukutin-related protein(FKRP). Additionally, mutations in the FKRP gene also cause limb-girdle muscular dystrophy type 2I(LGMD2I), a considerably milder allelic variant than MDC1C. All of these diseases are associated withsecondary changes in muscle a-dystroglycan expression. To elucidate the function of FKRP and fukutin andexamine the effects of MDC1C patient mutations, we have determined the mechanism for the subcellularlocation of each protein. FKRP and fukutin are targeted to the medial-Golgi apparatus through their N-terminiand transmembrane domains. Overexpression of FKRP in CHO cells alters the post-translational processingof a- and b-dystroglycan inhibiting maturation of the two isoforms. Mutations in the DxD motif in the putativeactive site of the protein or in the Golgi-targeting sequence, which cause FKRP to be inefficiently trafficked tothe Golgi apparatus, did not alter dystroglycan processing in vitro. The P448L mutation in FKRP that causescongenital muscular dystrophy changes a conserved amino acid resulting in the mislocalization of themutant protein in the cell that is unable to alter dystroglycan processing. Our data show that FKRP andfukutin are Golgi-resident proteins and that FKRP is required for the post-translational modification ofdystroglycan. Aberrant processing of dystroglycan caused by a mislocalized FKRP mutant could be a novelmechanism that causes congenital muscular dystrophy.

INTRODUCTION

The congenital muscular dystrophies (CMDs) are a hetero-geneous group of disorders that are characterized by early onsetmuscular dystrophy (reviewed in 1,2). Several of thesedisorders are also associated with brain defects, mentalretardation and abnormalities of the visual system (1–3).Recently, the genes for some of these diseases have been iden-tified (4–6). One of the most severe of these disordersis Fukuyama congenital muscular dystrophy (FCMD), whichis caused by mutations in the gene encoding fukutin. FCMD isthe second most common muscular dystrophy in the Japanesepopulation and predominates in populations originating fromNorth East Asia (7). FCMD patients are more severely affectedthan those with Duchenne muscular dystrophy (DMD) and oftennever walk. These patients are mentally retarded, affected by

epilepsy and have severe morphological brain abnormalitiesindicative of a defect in cortical neuronal migration (8). Themajority of FCMD patients have inherited the same mutation,namely the insertion of a retrotransposon in the 30 untranslatedregion of the FCMD gene (4). Compound heterozygotes havealso been described that have point mutations in one alleleof the FCMD gene and the retrotransposon on the secondallele (9). However, no patients have been described that arehomozygous for the point mutation suggesting that thecomplete lack of fukutin could result in embryonic lethality (9).The FCMD gene was predicted to encode a secreted proteincalled fukutin. However, due to the lack of suitable antibodies,the subcellular location of fukutin has not been determined (4).Interestingly, muscle biopsies from FCMD patients showa dramatic reduction in glycosylated a-dystroglycan, a com-ponent of the dystrophin protein complex (see 10 for a

*To whom correspondence should be addressed at: Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.Tel: þ44 01865271860; Fax: þ44 01865271853; Email: [email protected]{Present address: National Blood Service, John Radcliffe Hospital, Oxford, OX3 9DU, UK.{The authors wish it to be known that, in their opinion, these two authors should be considered as joint First Authors.

# 2002 Oxford University Press Human Molecular Genetics, 2002, Vol. 11, No. 26 3319–3331

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 2: Functional requirements for fukutin-related protein in the Golgi

Figure 1. Sequences and expression of murine FKRP and fukutin. The sequences of mouse FKRP (A) and fukutin (B). The underlined sequence corresponds to thehydrophobic N-terminal sequence that forms the Golgi signal anchor of FKRP and fukutin. The residues highlighted with bold lettering are the proposed active siteDxD of many glycosyltransferases (25). The tissue distribution of the FKRP and fukutin transcripts was determined on northern blots of mRNAs isolated fromdifferent mouse tissues (C). The 2.8 kb FKRP transcript is widely expressed with highest levels in brain, lung, heart kidney and liver (upper panel). The 6.8 kbfukutin transcript is detected predominantly in brain, liver and kidney (middle panel). The same blot was re-hybridized with a b-actin cDNA to show that similarlevels of mRNA were loaded (lower panel).

3320 Human Molecular Genetics, 2002, Vol. 11, No. 26

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 3: Functional requirements for fukutin-related protein in the Golgi

Figure 1 continued.

Human Molecular Genetics, 2002, Vol. 11, No. 26 3321

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 4: Functional requirements for fukutin-related protein in the Golgi

recent review), suggesting a link between FCMD, DMD anddystroglycan processing (11,12).

A second disorder, MDC1C is caused by mutations in thegene encoding a homologue of fukutin called fukutin-relatedprotein (FKRP) (13). MDC1C patients have severe musculardystrophy, however only a subset of these patients have braininvolvement (14). A milder allelic disorder LGMD2I is alsocaused by mutations in the FKRP gene (15,16). A wide rangeof mutations have been described in patients with MDC1C andLGMD2I however, in common with FCMD, patients with twoputative null alleles have not been described. Patients withMDC1C and LGMD2I also have abnormally processeda-dystroglycan that has a lower relative molecular mass andabundance suggesting that in common with fukutin, FKRP maybe involved in protein modifications such as glycosylation(13,17). However, MDC1C and LGMD2I patients havedetectable levels of glycosylated a-dystroglycan as determinedby immunoblotting showing that mutations in the FKRP geneaffect dystroglycan processing differently when compared tothe apparent absence of glycosylated dystroglycan in patientswith FCMD (13,16).

Direct evidence supporting a role for defects in proteinglycosylation causing CMD-like disorders comes from cloningof the genes mutated in Muscle–eye–brain (MEB) diseaseand the myodystrophy (myd ) mouse (6,18). MEB is similarto FCMD and is caused by mutations in the geneencoding O-mannose beta-1,2-N-acetylglucosaminyltransferase(POMGnT1) (6). This enzyme is involved in the synthesis ofO-mannosyl glycans and is expressed at its highest levels inmuscle and heart. MEB patients have a secondary deficiency ina-dystroglycan suggesting that dystroglycan may be a substratefor POMGnT1 (19). The myd mouse is the only naturallyoccurring model of CMD and is caused by a deletion in the geneencoding the putative glycosyltransferase LARGE (18). Inaddition to muscular dystrophy, these mice also have sensor-ineural hearing loss suggesting some degree of central nervoussystem involvement (18). Muscle from this mouse also hasincorrectly processed a-dystroglycan further implicating a rolefor protein glycosylation in the congenital muscular dystrophies.

Recently, it was shown that a-dystroglycan purified frommuscle samples from FCMD and MEB patients and the mydmouse has a molecular weight of 90 kDa and is only detectedby antibodies raised against the core of a-dystroglycan (20).Furthermore, a-dystroglycan derived from these sources had asignificantly reduced ability to bind to laminin, neurexin andagrin (20). These studies show that post-translation disruptionof the dystroglycan:ligand interface could cause CMD-likechanges in muscle. In a parallel study, it was shown thatdepletion of brain dystroglycan causes a neuronal migrationabnormality that is similar to that seen in the myd mouse andin patients with FCMD and MEB (21). Together, theseinteresting findings show that post-translation modification ofa-dystroglycan is required for normal brain development.However, the precise role of fukutin, POMGnT1 and LARGEremains to be formally established.

To determine the function of FKRP we investigated themechanism for the subcellular localisation of the protein andexamined its role in dystroglycan processing. For comparativepurposes we also determined the subcellular location offukutin. We have shown that both proteins are targeted to the

Golgi apparatus by their N-termini and transmembranedomains. We also show that FKRP has a direct effect ondystroglycan processing in vitro. Modelling of the mutationP448L found in a consanguineous MDC1C family (13)revealed that the mutant protein is mislocalized in the celland consequently is unable to alter the processing ofdystroglycan. Our data support the idea that FKRP and fukutinare involved in protein modification and suggest that abnormaldystroglycan processing is involved in the pathogenesis ofMDC1C and FCMD.

RESULTS

Cloning and expression analysis of FKRP and fukutin

cDNA clones encoding the mouse orthologues of FKRP andfukutin were isolated from a mouse brain library using genespecific PCR probes. The 2.8 kb murine FKRP cDNA encodesa protein of 494 amino acids and has 94% sequence identity toits human orthologue (Fig. 1A). The mouse fukutin cDNAencodes a protein of 461 amino acids and shares 90% sequenceidentity with its human orthologue (Fig. 1B). These cDNAclones were used to produce the expression constructsdescribed below (Table 1). The high degree of primarysequence similarity between the human and murine orthologuesof FKRP and fukutin facilitates the use of these mouse genesfor generating in vitro (see later) and in vivo models forMDC1C and FCMD.

For direct comparison, the tissue distribution of the mouseFKRP and fukutin transcripts were determined by hybridisingthe same northern blot with part of the cognate cDNA. TheFKRP transcript is 2.8 kb and is found at highest levels inthe brain, lung, heart, kidney and liver (Fig. 1C, upper panel).The fukutin transcript is 6.8 kb and is detected predominantlyin brain, liver and kidney (Fig. 1C, middle panel). Both of thetranscripts are expressed in the same tissues at similar levelsincluding the heart and skeletal muscle. The blot was strippedand re-hybridized with b-actin cDNA to demonstrate equalloading of mRNA (Fig. 1C, lower panel).

Sub-cellular localisation of FKRP and fukutin

Hydrophobicity plots and secondary structure analysispredict that FKRP and fukutin are type II membrane proteins(N-terminus outside, C-terminus inside). To determine thesubcellular location of FKRP and fukutin, several different celllines were transfected with expression constructs encodingeach protein (Table 1). Myc-tagged FKRP co-localizedprecisely with a-mannosidase II identifying the medial-Golgiapparatus in NRK cells (Fig. 2A). Similar results were obtainedwith fukutin indicating that both proteins are Golgi-residents(Fig. 2A). Comparison of myc-tagged FKRP with untaggedFKRP showed that the addition of an epitope tag at theC-terminus of the protein had no apparent effect on the locationof FKRP (data not shown). Treatment of the cells withbrefeldin A (BFA) or nocodazole (NC) resulted in disassemblyof the Golgi apparatus and the redistribution of each proteininto the endoplasmic reticulum (BFA) or into discrete clusters(NC, data not shown) indicative of Golgi-resident proteins.

3322 Human Molecular Genetics, 2002, Vol. 11, No. 26

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 5: Functional requirements for fukutin-related protein in the Golgi

FKRP and fukutin are predicted to be phospholigandtransferases and are possibly glycosyltransferases (13,17).Many glycosyltransferases are Golgi-residents and are main-tained in the organelle using a variety of different strategies. Todetermine the mechanism that targets FKRP and fukutin to theGolgi apparatus, several constructs were produced thatcontained part of each protein fused to green fluorescentprotein (GFP). The N-terminal 33 amino acids of FKRP aresufficient to recruit the GFP chimera to the Golgi apparatus(Fig. 2B). This region contains the transmembrane domain withthe N-terminal amino acids in the cytoplasm. Similar resultswere obtained with the N-terminus of fukutin fused to GFP(Fig. 2B). These data suggest that both proteins are targetedto the Golgi apparatus by a non-cleavable signal anchorsequence. The remaining lumenal domain of FKRP fused to theC-terminus of GFP was not targeted to the Golgi apparatus andremained diffuse within the cytoplasm of the cell and excludedfrom the nucleus (Fig. 2B). The corresponding region offukutin again behaves identically to the lumenal region ofFKRP in that it remains in the cytoplasm of the cell (Fig. 2B).In control experiments the two empty GFP vectors were nottargeted to the Golgi apparatus but were found in the cytoplasmand nucleus of transfected NRK cells (data not shown).

Several Golgi-resident proteins are recruited to the Golgiapparatus by a process known as kin recognition that involvesthe stem region of the protein (see 22 for a review). The stemlies between the transmembrane domain and catalytic domainof Golgi-resident glycosyltransferases. To determine whetherFKRP and fukutin could be targeted to the Golgi apparatusby kin-recognition, FK-pCIneo was co-transfected withGFP-FKRP-CT (Fig. 2C). GFP-FKRP-CT encodes the entireC-terminus of FKRP and will only be targeted to the Golgiapparatus through a potential association with fukutin. COS-7cells expressing both proteins were examined for co-localisation.FK-pCIneo is efficiently targeted to the Golgi apparatus butwas unable to recruit the C-terminal FKRP-GFP construct tothe same site (Fig. 2C). Similarly, FK-CT was not recruited tothe Golgi apparatus following co-transfection with full lengthFKRP (data not shown). These data show that FKRP andfukutin are targeted to the Golgi apparatus through theirN-termini and transmembrane domains and are unable torecruit each other to the organelle by kin association.

Hydrophobicity and secondary structure analysis was extendedto the other similar proteins implicated in congenital musculardystrophy (fukutin, LARGE, POMGnT1). Alignment of theN-terminal and transmembrane domains of each of these proteinsidentified a conserved motif R/KxxR/K immediately adjacent tothe transmembrane domain. These basic residues are alsoconserved in several other Golgi-resident proteins such asa-mannosidase-II and beta4GalTI (Fig. 3A). To examine theeffect of the conserved amino acids on the location of FKRP bothof the arginine codons (R2 and R5) were mutated to glutamic acidcodons. These mutations were placed in two different backbones;the first was in the context of full length FKRP, the second inthe NT-FKRP-GFP Golgi-targeted construct. The polypeptidesexpressed by both constructs are localized to the perinuclearregion but are not retained in, or targeted to, the medial part of theGolgi apparatus (Fig. 3B). It can therefore be concluded that theconserved basic residues in the N-terminus of FKRP are requiredfor Golgi-targeting and/or retention.

Expression of FKRP alters dystroglycan processingin vitro

One of the salient features of MDC1C and FCMD are thesecondary changes in a-dystroglycan (12,13). It has beenpostulated that these qualitative differences in a-dystroglycanare caused by an abnormality in dystroglycan modification.To determine the effect of FKRP on the processing of a-dystroglycan CHO cells were co-transfected with dystroglycanand FKRP expression constructs and mutants thereof (Table 1).CHO cells were chosen because they have previously beenshown to produce highly glycosylated and correctly processeda- and b-dystroglycan (23). In these experiments we used achick dystroglycan expression construct (ChDG) and anti-bodies specific for the chick core protein. This experimentalparadigm allowed us to examine the effects of FKRP ondystroglycan processing without using antibodies againstglycosylated epitopes. We also used FKRP without theepitope tags because this could potentially interfere withthe function of FKRP. Furthermore, using antibodies specific tothe a-dystroglycan core protein and 9E10 that detects a mycepitope tag placed at the C-terminus of b-dystroglycan, wewere able to identify the different dystroglycan isoforms

Table 1. Constructs used in this study

Name Vector Description

FKRP pCIneo Full length mouse FKRP cdsFKRP-myc pCIneo Full length FKRP with a C-terminal myc tag.FK pCIneo Full length mouse fukutin cdsNT-FKRP-GFP pEGFP-N3 aa 1–33 of FKRP fused to the N-terminus of GFPGFP-FKRP-CT pEGFP-C2 aa 29–end fused to the C-terminus of GFPNT-FK-GFP pEGFP-N3 aa 1–36 of fukutin fused to the N-terminus of GFPGFP-FK-CT pEGFP-C2 aa 36–end of fukutin fused to the C-terminus of GFPChDG pCMV-Tag1 Full length chick dystroglycanFKRP-NNN pCIneo Full length FKRP with aa 362–364 mutated from DVD to NNNFKRP-R2E, R5E pCIneo Full length FKRP with aa 2 and 5 mutated from R to EFKRP-P448L pCIneo FKRP with aa 448 mutated from P to LFKRP-myc-P448L pCIneo FKRP-myc with aa 448 mutated from P to L

cds: Coding sequence; aa: amino acids.

Human Molecular Genetics, 2002, Vol. 11, No. 26 3323

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 6: Functional requirements for fukutin-related protein in the Golgi

Figure 2. Golgi-targeting of FKRP and fukutin. Golgi localization of FKRP and fukutin (A). Transfected NRK cells were stained for FKRP-myc (upper panel,green), fukutin (lower panel, green) and a-mannosidase II (manII, middle panels, red). The merged images show precise co-localization of FKRP and fukutin witha-mannosidase II. Golgi targeting sequences in FKRP and fukutin (B). NRK cells transfected with the GFP constructs shown on the left of the figure were stainedwith an anti a-mannosidase II antibody to identify the medial part of the Golgi apparatus. Two channel confocal images were captured to show the location of theGolgi apparatus (red channel, a-mannosidase II) and the different GFP constructs as indicated (green channel). The merge images are shown in the right handpanels. Only the N-terminal constructs, NT-FKRP-GFP and NT-FK-GFP are targeted to the Golgi apparatus. No targeting is seen with the constructs containingthe lumenal domains of FKRP and fukutin (GFP-FKRP-CT and GFP-FK-CT) or with the two empty GFP vectors (pEGFP-C2 and pEGFP-N3) used in this experi-ment (data not shown). Testing kin recognition between FKRP and fukutin (C). COS-7 cells transfected with GFP-FKRP-CT and full length fukutin were examinedfor co-localization of the two antigens. GFP-FKRP-CT stains punctae in the cytoplasm whereas fukutin is restricted to the Golgi apparatus. There is no apparentco-localization of the two antigens suggesting that fukutin cannot recruit FKRP to the Golgi apparatus. Scale bar¼ 20mm.

3324 Human Molecular Genetics, 2002, Vol. 11, No. 26

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 7: Functional requirements for fukutin-related protein in the Golgi

originating from cleavage and processing of the precursorprotein.

We also compared the processing of dystroglycan with twoFKRP mutants. FKRP-NNN has a mutation of residues 362 to364 such that the sequence DVD is mutated to NNN. The DxDmotif in FKRP is indicative of enzymatic function and is highlyreminiscent of motifs found in many other inverting and non-inverting transferases that add sugars to other sugars, phosphatesand proteins (24–26). Mutations of the DxD motif to NNN havebeen used to abolish the function of many putative glycosyl-transferases (see 27 for example). The other mutant assayed inour expression system was FKRP-R2E, R5E that is poorlytargeted to, or retained in, the Golgi apparatus (Fig. 3).

Expression of ChDG alone in CHO cells produces dystrogly-can that is processed into three major proteins, b-dystroglycan(43 kDa), a-dystroglycan (160 kDa) and an a-dystroglycanisoform of 90 kDa (Fig. 4A and B). The 90 kDa and 160 kDaproteins are not detected with 9E10 showing that they aredifferentially processed forms of a-dystroglycan (data notshown). Co-expression of ChDG and FKRP alters the patternof dystroglycan immunoreactivity such that there is a markeddecrease in the levels of 160 kDa dystroglycan and an increase inthe 90 kDa isoform and the appearance of an a-dystroglycancross-reactive protein of 60 kDa (Fig. 4A and B). Interestingly,FKRP also alters the processing of b-dystroglycan increasing theabundance of a smaller b-dystroglycan isoform of 41 kDa(Fig. 4A and C). We then examined the effect of two FKRP

mutants on dystroglycan processing. Both mutants behave in asimilar manner by reducing the levels of the 90 kDa dystrogly-can (compared to the expression of wild-type FKRP) but leavingthe levels of 160 kDa dystroglycan apparently unaltered (Fig. 4Aand B). Thus it can be concluded that each mutant is unable toaffect the processing of dystroglycan in vitro. In controlexperiments the presence of the FKRP transcript in each of theappropriate transfections was demonstrated by RT–PCR(Fig. 4D). Each of these experiments was performed at leastthree times with a similar outcome. Two antibodies against eachdystroglycan isoform were also used to ensure that each bandwas correctly assigned. Antibodies VIA41 and IIH6 thatrecognise glycan-dependent epitopes on human dystroglycandid not cross-react with chick dystroglycan.

Comparative analysis of dystroglycan in the presence andabsence of FKRP by two dimensional gel electrophoresisshowed an increase in the abundance of low molecular weighta-dystroglycan isoforms (Fig. 4E). The pattern of dystroglycanimmunoreactivity on 2D gels detected with an antibody thatrecognises the core protein shows a major broad band of160 kDa, indicative of heterogeneous glycosylation, with a lowpI (Fig. 4E). Other weaker bands can also be seen thatcorrespond to the 90 kDa a-dystroglycan isoform. In cellstransfected with FKRP and ChDG, three major proteins of160 kDa, 90 kDa and 60 kDa are detected (Fig. 4E). Expressionof FKRP results in a dramatic reduction in the levels of160 kDa a-dystroglycan when compared to cells only expressing

Figure 3. Mutational analysis of the Golgi-targeting sequence in FKRP. (A) Sequence alignment of the cytoplasmic N-terminus and transmembrane domains ofFKRP, fukutin, LARGE, POMGnT1, a-mannosidase II and beta4GalT I identifies a pair of conserved basic amino acids (asterisks) immediately adjacent to thetransmembrane. Both arginine codons were mutated to glutamic acid in FKRP (mut-FKRP). COS-7 cells transfected with the mutant (R2E, R5E) and wild-typeFKRP (red) show that the mutant is not precisely targeted or retained by the Golgi apparatus (green) but labels diffusely around the Golgi stack and the nuclearmembrane. By contrast wild-type FKRP precisely co-localizes with marker at the marker apparatus. Scale bar¼ 20 mm.

Human Molecular Genetics, 2002, Vol. 11, No. 26 3325

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 8: Functional requirements for fukutin-related protein in the Golgi

ChDG (Fig. 4E). In the cells expressing FKRP and ChDG thelower molecular weight a-dystroglycan cross-reactive proteinspredominate (asterisks in Fig. 4E). The 90 kDa and 60 kDadystroglycan immunoreactive proteins do not appear to beheterogeneous since they have a discrete isoelectric point (cf160 kDa a-dystroglycan Fig. 4E). It is possible that they areunderglycosylated proteolytic fragments since the unmodifiedcore dystroglycan has a predicted relative molecular mass of71.5 kDa. These data support our findings that expression ofFKRP causes an alteration in dystroglycan processing in vitro.

Functional analysis of the P448L mutation in theFKRP gene

Given that our experimental system has allowed us to analysethe localization and presumed function of FKRP, we used

this-paradigm to model some of the mutations seen inMDC1C patients. For our initial studies we chose mutationsidentified in severely affected patients. The mutation P448Lalters a highly conserved amino acid in the C-terminus ofFKRP. Proline appears to be invariant at this position invertebrate FKRPs, insect FKRPs, a distant bacterialhomologue Ricketssia prowazekii, RP-689 and fukutin(Fig. 5A). FKRP-P448L localised to the nuclear membranewith a discrete but diffuse perinuclear extension (Fig. 5B). Themutant fails to precisely co-localise with the Golgi marker,suggesting that it is inefficiently trafficked or retainedin the Golgi apparatus (Fig. 5B). When this mutant isco-transfected with ChDG in CHO cells it has noapparent effect on the processing of dystroglycan (Fig. 5C).The pattern of dystroglycan immunoreactive proteinsdiffers considerably from the control transfections with

Figure 4. FKRP affects dystroglycan processing. CHO cells were transfected with ChDG (lane 1), ChDG and FKRP (lane 2) ChDG and FKRP-NNN (lane 3) andChDG and FKRP-R2E, R5E (lane 4). No dystroglycan cross-reactive proteins were detect in untransfected CHO cells. Western blots of cell extracts were probed withan antibody against a-/b-dystroglycan (A), a-dystroglycan (B) or the myc epitope tag located at the C-terminus of b-dystroglycan (C). The asterisks mark the positionsof the different isoforms of a- and b-dystroglycan. An RT–PCR assay for FKRP expression was performed on the transfected CHO cells (D). FKRP is detected in lanes2–4 but not in lane 1 or the lane with no template (nt). 2D–PAGE analysis of dystroglycan produced in CHO cells (E). CHO cells were transfected as indicated.Proteins extracted from the cells were analysed by 2D–PAGE. In the first dimension proteins were separated by isoelectric point whilst the second dimension separatesproteins by relative molecular mass. a-dystroglycan, detected with the Sheep 1 antibody, appears as a broad major band in cells expressing ChDG. In cells expressingFKRP the 160 kDa a-dystroglycan band is reduced whilst two partially processed intermediates of 90 kDa and 60 kDa (asterisks) are found.

3326 Human Molecular Genetics, 2002, Vol. 11, No. 26

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 9: Functional requirements for fukutin-related protein in the Golgi

ChDG and FKRP and is indistinguishable from the dystro-glycan profile of cells only transfected with ChDG(Fig. 4C). Interestingly the P448L-FKRP mutant is remini-scent of the R2E, R5E-FKRP mutant (see Fig. 3 forcomparison) in that it is not properly targeted to the Golgiapparatus and has no apparent effect upon the processing ofdystroglycan. These data suggest that the FKRP-P448L ismistargeted in the cell and therefore cannot alter the processingof a-dystroglycan.

DISCUSSION

In this paper we have shown that FKRP and fukutin are Golgi-resident proteins and that the N-terminus and transmembranedomain of each protein is sufficient to define their subcellularlocation. Using an in vitro expression system we also show thatFKRP overexpression alters the processing of dystroglycan butthat several mutants, including the mutation P448L that resultsin a severe form of muscular dystrophy are unable to modify

Figure 5. The P448L mutation is inefficiently trafficked to the Golgi apparatus. Sequence alignment of the region of FKRP at proline 448. P448 (asterisk) isconserved in a variety of different FKRP orthologues and also in fukutin. Dr, Danio rerio; Dm, Drosophila melanogaster; Ag, Anopheles gambiae;Rp, Rickettsia prowazekii; Mm, Mus musculus; Ci, Ciona intestinalis. COS-7 cells transfected with FKRP-P448L show that the mutant protein does not preciselyco-localize with the Golgi marker (B). FKRP-P448L immunoreactivity is spread diffusely around the region around the Golgi apparatus and is also seen at thenuclear membrane. Scale bar¼ 20mm. Extracts were prepared from CHO cells transfected with ChDG (lane 1), ChDG and FKRP (lane 2), ChDG andFKRP-P448L (lane 3) and western blotted with an antibody against a-/b-dystroglycan and a-dystroglycan (C). The mutant protein does not produce any discernibledifference in dystroglycan when compared to cells only expressing ChDG. By contrast, expression of wild-type FKRP produces a markedly different pattern ofimmunoreactive proteins. The asterisks mark the positions of the different isoforms of a- and b-dystroglycan.

Human Molecular Genetics, 2002, Vol. 11, No. 26 3327

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 10: Functional requirements for fukutin-related protein in the Golgi

a-dystroglycan. These data strongly support our initial proposalthat FKRP and fukutin are proteins that are required for thepost-translational modification of dystroglycan (5,16).

It is now clear that several forms of congenital musculardystrophy are associated with secondary alterations ina-dystroglycan processing. These abnormalities are seen inpatients with MDC1C, FCMD, MEB and in the myd mouse.Bio-informatic clues about the function of the genes that causethese disorders and the secondary changes in a-dystroglycan inthe muscle suggest that each of these proteins is potentially aglycosyltransferase. However, thus far only POMGnT1 hasbeen shown to directly catalyse a glycosylation reaction and inno case has a direct effect on dystroglycan been demonstrated.Transfection of genes encoding glycosyltransferases into cellsand subsequent analysis of their glycome is a powerful newapproach to defining substrates and activities for glycosyl-transferases with unknown activities (28). We have used avariation of this strategy to examine the processing ofdystroglycan in CHO cells in the presence of FKRP.Overexpression of FKRP clearly has an effect on dystroglycanmaturation such that there is a reduction in the 160 kDa isoformof a-dystroglycan and a concomitant increase in 90 kDa and60 kDa a-dystroglycan cross-reactive proteins (Fig. 4). Whilstthe molecular identities of the 90 kDa and 60 kDa dystroglycanisoforms are unknown, these proteins could represent imma-ture processing intermediates or proteolytic fragments ofa-dystroglycan (see below). These changes in dystroglycanprocessing appear to be a direct consequence of functionalFKRP since the three mutants that were constructed had littleor no effect on dystroglycan processing in vitro (Fig. 4 andFig. 5). Our studies provided the first demonstration that FKRPhas a direct effect on dystroglycan processing in vitro.

Whilst the precise activity of FKRP and fukutin is unknownwe have been able to show that FKRP directly affectsdystroglycan processing. The downward shift in relativemolecular weight of a-dystroglycan is possibly explained bythe addition of terminating glycans to the dystroglycanbackbone such that no additional modifications can be madeor by altering the stability of dystroglycan rendering itsusceptible to endogenous proteases. It is well-documentedthat glycosylation increases the stability of proteins in theextracellular matrix (ECM) and in plasma as well as having arole in receptor:ligand interactions (29). Thus overexpressionof FKRP could render dystroglycan sensitive to matrix orsecreted proteases. It is also interesting to note that FKRP alsoaffects b-dystroglycan processing again reducing the relativemolecular weight of the protein. This effect could again beexplained by proteolysis or incomplete glycosylation ofb-dystroglycan.

It still remains to be established whether FKRP, fukutin andLARGE have any catalytic activity. However, by comparisonto POMGnT1 each protein may be involved in O-linkedmannosylation. Several proteins in yeast are required forO- and N-linked mannosylation (30). Of these the proteins,Mnn4p has significant primary sequence similarity to FKRP. S.cerevisiae strains with mutations in the MNN4 gene havereduced mannosylphosphate transferase activity and reductionin mannosylphosphate content of cell wall mannan (31,32). Nocatalytic activity has been ascribed to Mnn4p, however ithas been proposed that Mnn4p regulates the levels or activity

of other enzymes in the mannosylation pathway (30,32).Therefore it has been suggested that MNN4 in yeast has adominant (positive) regulatory role (30). Given that a yeasthomologue of FKRP appears to be a non-catalytic regulator ofmannosylation, it is formally possible that FKRP has such arole in humans.

Several studies have shown that a-dystroglycan is a heavilysialylated mucin-like glycoprotein that is found in differentvariants that are formed by tissue-specific glycosylation (forreview see 33,34). Dystroglycan glycosylation is important forbinding to laminin and agrin in the basal lamina (35–37).Abnormalities in the basal lamina have also been described inthe muscles and brains of FCMD patients. These defectsinclude reduction in the levels of several components of themuscle and neuronal ECM and alterations in immunoreactivityof members of the dystrophin-associated protein complex(11,38). Furthermore, alterations in laminin-a2 immuno-reactivity have been described in the muscles of patients withFCMD (38,39). Thus, FCMD may result from defects inthe ECM or basal laminae that could involve components of thedystrophin-associated protein complex (40). Alterations in theprocessing of a-dystroglycan could have several importanteffects on basal lamina formation and function. By analogy toFCMD, patients with MDC1C could have similar basal laminadefects. One possible explanation for the severe musculardystrophy in these patients is that mutations that abolish thefunction of FKRP produce a-dystroglycan that has a reducedcapacity to bind to extracellular laminin and thus disrupts thebasal lamina of the muscle (Fig. 6). Consistent with thishypothesis, MDC1C patients often have secondary changes inlaminin immunoreactivity (16). The simplest explanation forthe phenotype of patients with MDC1C is that mutations inGolgi-resident FKRP cause aberrant or incomplete glycosyla-tion of a-dystroglycan and possibly other ECM proteins suchthat dystroglycan has a reduced capacity to bind the ECMcomponents effectively weakening the bridge between the DPCand the ECM (Fig. 6). For diseases such as FCMD and MEBand in the myd mouse it has been shown that low molecularweight a-dystroglycan has a reduced affinity for several ECMproteins supporting the model presented in Figure 6 (20).

In this paper we have determined the mechanism for Golgi-targeting and retention of FKRP. Both FKRP and fukutin aretargeted to the Golgi apparatus through their N-termini andtransmembrane domains. Both proteins co-localise with themedial Golgi marker a-mannosidase II. We have shown that inaddition to the transmembrane domain of FKRP the N-terminalarginine residues play an important role in Golgi-targeting and/or retention. b-1,4-galactosyltransferase is also targeted toGolgi apparatus via its membrane-spanning domain (41).However, single cysteine and histidine residues within thetransmembrane domain are required for oligomerization andGolgi-retention (42). The transmembrane domain of FKRPdoes not have a cysteine or histidine residue suggesting thathomo-oligomerization is unlikely to be a retention mechanismfor FKRP. Furthermore, we have shown that the N-terminalarginine residues of FKRP are important determinants forGolgi-targeting or retention. This pair of basic residues is foundin many medial Golgi-residents, including those implicated inCMD (Fig. 3), and may govern the Golgi-localization of asubset of Golgi-resident proteins. We have also shown that the

3328 Human Molecular Genetics, 2002, Vol. 11, No. 26

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 11: Functional requirements for fukutin-related protein in the Golgi

FKRP mutant P448L is poorly targeted to or retained in the Golgiapparatus (Fig. 5). Given that the N-terminus of FKRP is criticalfor Golgi targeting (Fig. 3), the FKRP-P448L mutant protein maybe inefficiently retained targeted to, or retained in, the Golgiapparatus because it is misfolded. These data demonstrate theimportance for the Golgi localization of FKRP and identifyseveral sequence elements that are required for this process.

In summary, we have shown that FKRP and fukutin areGolgi-resident proteins that are targeted to the organelle using anon-cleavable signal anchor. FKRP directly affects theprocessing and/or maturation of dystroglycan in vitro and thatthis function is perturbed by several different mutations in thegene. We have also demonstrated a functional requirement forFKRP targeting to the Golgi apparatus such that the MDC1Cpatient mutation, P448L, is mistargeted and unable to affectdystroglycan processing. These data provide the first functionalcharacterization of FKRP and offer a potential molecularmechanism to explain the muscle disease in MDC1C patients.These studies will also shed light on the other CMDs that areassociated with secondary changes in a-dystroglycan.

MATERIALS AND METHODS

Cloning, expression constructs and northern blotting

Mouse cDNA clones encoding FKRP and fukutin were isolatedfrom a normalised mouse brain cDNA library cloned in pPC86(Invitrogen) by colony hybridization and sequenced usingstandard methods. The EMBL accession numbers for eachsequence are AJ511806 (FKRP) and AJ511807 (fukutin). Thecomplete FKRP cDNAwas subcloned into the NotI site of pCIneo(Promega). Golgi-targeted GFP constructs were made by PCRamplification of the regions encoding the first 33 amino acids ofFKRP with the primers, FKRPgfpf (50-TGGAATTCTCAA-GGTGAAACCACTCTTCTTG) and FKRPgfpr (50-TGGGAT-CCGTTTCTGGGCTGGTGTTGTAG) and the first 36 aminoacids of fukutin with the primers, FKgfpf, (50-TGAATTCTAT-TCTGTTTACATGAGAAGATGAA) and FKgfpr, (50-TGGG-ATCCATTCCTAGACAAATAGTGCTT). PCR products were

purified and ligated into the EcoRI/BamHI sites of pEGFP-N3(Clontech). The remainder of the FKRP cDNA, lacking the regionencoding the N-terminus and transmembrane domain, was ampli-fied by PCR using the primers, FKRPDgf (50-CCGAATTCC-ACCAGCCCAGAAACTCCCG) and FK2Dgr (50-CGCTCGA-GAACCGCCTGTCAAGCTTAAGAG) and subcloned intopEGFPC2 (Clontech). The remainder of the fukutin cDNAencoding amino acids 36–461 was PCR amplified using theprimers FKDgf (50-TGGAATTCGGACCGGGTTCATCAA-AATC) and FKDgr (50-TGGGTACCCTGAGCTTAGTTTTCT-CAGTAC) and subcloned into pEGFPC2. A myc epitope tag wasintroduced at the C-terminus of FKRP by ligating an annealedoligonucleotide duplex; FKRPmycF (50-AGCTTGACAGGCG-GTGAGCAAAAGCTCATTTCTGAAGAGGACTTGTGAGG-TAC) and FKRPmycR (50-CTCACAAGTCCTCTTCAGAAA-TGAGCTTTTGCTCACCGCCTGTCA) into the HindIII/KpnIsites at the 30 end of the FKRP coding sequence. All clones wereverified by sequencing. Northern blots were purchased fromOrigene (Origene Technologies, Rockville, MD) and hybridizedwith the mouse FKRP and fukutin cDNAs in Rapid-hyb buffer(Amersham-Pharmacia), according to the manufacturers instruc-tions. RT–PCR was performed on RNA extracted from CHO cellstransfected with FKRP expression constructs and derivativesthereof using standard protocols.

Site-directed mutagenesis

Site-directed mutagenesis was performed on plasmid DNA(10 ng) using Pfu Turbo DNA polymerase (Stratagene) in a50ml volume with 125 ng of each primer and 0.2 mM dNTPs.PCR cycles were as follows: 95�C for 30 s followed by 18 cyclesof 95�C for 30 s, 55�C for 1 min, 68�C for 16 min. The mutatedDNA template was digested with DpnI (10 U) to remove theparental plasmid and used to transform XL1-Blue competentcells. Plasmid DNA was extracted from transformed XL1-Bluecultures using the Qiagen EndoFree Maxiprep kit according to themanufacturer’s instructions. Mutations were confirmed by DNAsequencing. The primers used to make the different mutants were:

FKRP-NNNf, 50-CCT TGG GAC TAC AAC AAC AAC CTGGGC ATC TAC C

Figure 6. A potential mechanism for congenital muscular dystrophy. The schematic shows how mutations in the FKRP gene could cause muscular dystrophy byinterfering with the processing of dystroglycan. The most likely affect of FKRP and fukutin mutations is to change the glycosylation of a-dystroglycan. This mayinterfere with the interactions of dystroglycan with laminin-2 and other ECM proteins or could reduce the stability of dystroglycan in the ECM.

Human Molecular Genetics, 2002, Vol. 11, No. 26 3329

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 12: Functional requirements for fukutin-related protein in the Golgi

FKRP-NNNr, 50-G GTA GAT GCC CAG GTT GTT GTT GTAGTC CCA AGGFKRP-448Lf, 50-G CAC TTC CTG CAG CTACTT GTC CCCCTG CFKRP-448Lfr, 50-G CAG GGG GAC AAG TAG CTG CAGGAA GTG CFKRP-2E5Ef, 50-GGC CCC ATG GAG CTC ACC GAG TGCTGG GCT GFKRP-2E5Er, 50-C AGC CCAGCACTC GGT GAG CTC CATGGG GCC

Altered residues are underlined.

Antibodies and immunocytochemistry

Polyclonal anti-FKRP antibodies (FKRP-FP) were producedin rabbits immunized with a thioredoxin fusion proteinspanning the entire lumenal domain of the protein (aminoacids 36–end). Polyclonal anti-fukutin antibodies (FK-CT)were produced in rabbits against a thioredoxin fusion proteinspanning the last 60 amino acids of mouse fukutin. Allantibodies were affinity purified and pre-absorbed againstthioredoxin by column chromatography using proteinscovalently attached to Sulfolink (Pierce and Warriner). Theanti-FKRP and fukutin antibodies only worked on recombi-nant protein produced by heterologous expression in mam-malian cells. Other groups have failed to produce anti-fukutinantibodies in spite of using multiple immunogens anddifferent strategies (4). The sheep anti-dystroglycan antibodieshave been described previously (43). Mouse anti-mannosidase IIwas purchased from BabCO (Berkley, CA) and the 9E10 anti-myc monoclonal antibody was purchased from Roche.Expression constructs were transfected into COS-7 (monkeykidney), NRK (normal rat kidney fibroblasts) and CHO(Chinese hamster ovary) cell lines using Fugene-6 (Roche)following the manufacturer’s instructions. For immunolocaliza-tion of FKRP and fukutin, cells grown on glass cover slips werefixed in 4% (w/v) paraformaldehyde in PBS at 4�C for 15 minand permeabilized in 0.1% (v/v) Triton X-100 in PBS for15 min at 4�C. Cells were extensively washed in PBS beforeapplying the antibodies. Antigens were detected by indirectimmunofluorescence with the following primary antibodies,FKRP-CT (diluted 1:25), FK-CT (diluted 1:100), anti-myc9E10 (diluted 1:200) and anti-a-mannosidase II (diluted1:10 000) followed by the secondary antibodies, rhodamine-Red X anti-rabbit-IgG (Jackson Immunoresearch) and Alexa488-labelled anti-mouse-IgG (Molecular Probes). Slides wereexamined by fluorescence microscopy using a Leica DMREmicroscope or by laser confocal microscopy using a Leica TCSSP2 microscope. Images were adjusted for brightness andcontrast using Adobe Photoshop version 6. For studies on theFKRP mutants, expression constructs were transfected intoCOS-7 cells as described above. The Golgi apparatus wasidentified by co-transfection of a Golgi-localized GFP construct(NT-FK-GFP). The antigens were identified by immunofluor-escence as described above. Slides were viewed with a Leicamicroscope and images were captured using a HamamatsuOrca ER CCD camera and OpenLab 3.0.9 software(Improvsion).

2D-gel electrophoresis and immunoblotting

Proteins were prepared from transfected CHO cells that werelysed with 2D rehydration sample buffer (8 M urea, 2% w/vChaps, 50 mM DTT, 0.2% v/v bio-lyte 3/10 ampholytes) andseparated by isoelectric focusing in the first dimension usingIPG strips and a Protean IEF Cell (BioRad) and SDS–PAGE inthe second dimension (10% Criterion gels, BioRad). Proteinswere transferred to a nitrocellulose membrane (BA83,Schleicher and Schuell) and probed with antibodies againstdystroglycan (diluted 1:1000) or the myc-epitope tag (diluted1:100). For western blots, protein samples were prepared forSDS–PAGE in SDS/urea buffer (4 M urea, 3.8% SDS, 20%glycerol, 75 mM Tris pH 6.8, 5% 2-mercaptoethanol) andimmunoblotted as described previously (44).

ACKNOWLEDGEMENTS

The authors thank Prof Kay Davies for support andencouragement, Dr Chris Ponting for helpful discussions,Ally Potter and Kara Hunter for help with cell culture andconfocal microscopy. This work was generously supported bygrants from the Wellcome Trust (D.J.B.), DeutscheForschungsgemeinschaft (DFG grant number Kr1039/7)(S.K.), The Muscular Dystrophy Campaign of Great Britainand Northern Ireland (F.M.) and The European Community(Myo-Cluster: GENRE grant QLG1 CT 1999 00870) (F.M.).D.J.B. is a Wellcome Trust Senior Fellow in Basic BiomedicalScience. M.A.B. is the recipient of a Wellcome Trust PrizeStudentship.

REFERENCES

1. Mercuri, E. and Muntoni, F. (2001) Congenital muscular dystrophies. InEmery, A.E.H. (ed.), The Muscular Dystrophies. Oxford University Press,Oxford, pp. 10–38.

2. Voit, T. (1998) Congenital muscular dystrophies: 1997 update. Brain Dev.,20, 65–74.

3. Blake, D.J. and Kroger, S. (2000) The neurobiology of Duchennemuscular dystrophy: learning lessons from muscle? Trends Neurosci., 23,92–99.

4. Kobayashi, K., Nakahori, Y., Miyake, M., Matsumura, K., Kondo-Iida, E.,Nomura, Y., Segawa, M., Yoshioka, M., Saito, K., Osawa, M. et al. (1998)An ancient retrotransposal insertion causes Fukuyama-type congenitalmuscular dystrophy. Nature, 394, 388–392.

5. Brockington, M., Blake, D.J., Brown, S.C. and Muntoni, F. (2002) The genefor a novel glycosyltransferase is mutated in congenital muscular dystrophyMDC1C and limb girdle muscular dystrophy 2I. Neuromuscul. Disord., 12,233–234.

6. Yoshida, A., Kobayashi, K., Manya, H., Taniguchi, K., Kano, H.,Mizuno, M., Inazu, T., Mitsuhashi, H., Takahashi, S., Takeuchi, M. et al.(2001) Muscular dystrophy and neuronal migration disorder caused bymutations in a glycosyltransferase, POMGnT1. Dev. Cell, 1, 717–724.

7. Eberhart, D.E. and Curran, T. (1998) Yayoi era mutation disrupts brain andmuscle [news]. Nat. Med., 4, 1002–1003.

8. Fukuyama, Y., Osawa, M. and Suzuki, H. (1981) Congenital progressivemuscular dystrophy of the Fukuyama type—clinical, genetic andpathological considerations. Brain Dev., 3, 1–29.

9. Kondo-Iida, E., Kobayashi, K., Watanabe, M., Sasaki, J., Kumagai, T.,Koide, H., Saito, K., Osawa, M., Nakamura, Y. and Toda, T. (1999) Novelmutations and genotype–phenotype relationships in 107 families withFukuyama-type congenital muscular dystrophy (FCMD). Hum. Mol.Genet., 8, 2303–2309.

10. Blake, D.J., Weir, A., Newey, S.E. and Davies, K.E. (2002) Function andgenetics of dystrophin and dystrophin-related proteins in muscle. Physiol.Rev., 82, 291–329.

3330 Human Molecular Genetics, 2002, Vol. 11, No. 26

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018

Page 13: Functional requirements for fukutin-related protein in the Golgi

11. Matsumura, K., Nonaka, I. and Campbell, K.P. (1993) Abnormalexpression of dystrophin-associated proteins in Fukuyama-typecongenital muscular dystrophy [see comments]. Lancet, 341,521–522.

12. Hayashi, Y.K., Ogawa, M., Tagawa, K., Noguchi, S., Ishihara, T.,Nonaka, I. and Arahata, K. (2001) Selective deficiency of alpha-dystroglycan in Fukuyama-type congenital muscular dystrophy. Neurology,57, 115–121.

13. Brockington, M., Blake, D.J., Prandini, P., Brown, S.C., Torelli, S.,Benson, M.A., Ponting, C.P., Estournet, B., Romero, N.B., Mercuri, E. et al.(2001) Mutations in the fukutin-related protein gene (FKRP) cause a formof congenital muscular dystrophy with secondary laminin alpha2 deficiencyand abnormal glycosylation of alpha-dystroglycan. Am. J. Hum. Genet., 69,1198–1209.

14. Topaloglu, H., Brockington, M., Yuva, Y., Talim, B., Haliloglu, G.,Blake, D., Torelli, S., Brown, S. and Muntoni, F. (2002) FKRP genemutations cause congenital muscular dystrophy 1C, mental retardation andcerebellar cysts. Neurology, in press.

15. Driss, A., Amouri, R., Ben Hamida, C., Souilem, S., Gouider-Khouja, N.,Ben Hamida, M. and Hentati, F. (2000) A new locus for autosomalrecessive limb-girdle muscular dystrophy in a large consanguineousTunisian family maps to chromosome 19q13.3. Neuromuscul. Disord., 10,240–246.

16. Brockington, M., Yuva, Y., Prandini, P., Brown, S.C., Torelli, S.,Benson, M.A., Herrmann, R., Anderson, L.V., Bashir, R., Burgunder, J.M.et al. (2001) Mutations in the fukutin-related protein gene (FKRP)identify limb girdle muscular dystrophy 2I as a milder allelic variant ofcongenital muscular dystrophy MDC1C. Hum. Mol. Genet., 10,2851–2859.

17. Aravind, L. and Koonin, E.V. (1999) The fukutin protein family-predicted enzymes modifying cell-surface molecules. Curr. Biol., 9,R836–R837.

18. Grewal, P.K., Holzfeind, P.J., Bittner, R.E. and Hewitt, J.E. (2001)Mutant glycosyltransferase and altered glycosylation ofalpha-dystroglycan in the myodystrophy mouse. Nat. Genet.,28, 151–154.

19. Kano, H., Kobayashi, K., Herrmann, R., Tachikawa, M., Manya, H.,Nishino, I., Nonaka, I., Straub, V., Talim, B., Voit, T. et al. (2002)Deficiency of alpha-dystroglycan in muscle-eye-brain disease. Biochem.Biophys. Res. Commun., 291, 1283–1286.

20. Michele, D.E., Barresi, R., Kanagawa, M., Saito, F., Cohn, R.D., Satz, J.S.,Dollar, J., Nishino, I., Kelley, R.I., Somer, H. et al. (2002) Post-translationaldisruption of dystroglycan-ligand interactions in congenital musculardystrophies. Nature, 418, 417–422.

21. Moore, S.A., Saito, F., Chen, J., Michele, D.E., Henry, M.D., Messing, A.,Cohn, R.D., Ross-Barta, S.E., Westra, S., Williamson, R.A. et al. (2002)Deletion of brain dystroglycan recapitulates aspects of congenital musculardystrophy. Nature, 418, 422–425.

22. Colley, K.J. (1997) Golgi localization of glycosyltransferases: morequestions than answers. Glycobiology, 7, 1–13.

23. Holt, K.H., Crosbie, R.H., Venzke, D.P. and Campbell, K.P. (2000)Biosynthesis of dystroglycan: processing of a precursor propeptide. FEBSLett., 468, 79–83.

24. Busch, C., Hofmann, F., Selzer, J., Munro, S., Jeckel, D. and Aktories, K.(1998) A common motif of eukaryotic glycosyltransferases is essential forthe enzyme activity of large clostridial cytotoxins. J. Biol. Chem., 273,19566–19572.

25. Wiggins, C.A. and Munro, S. (1998) Activity of the yeast MNN1alpha-1,3-mannosyltransferase requires a motif conserved in many otherfamilies of glycosyltransferases. Proc. Natl Acad. Sci. USA, 95,7945–7950.

26. Hagen, F.K., Hazes, B., Raffo, R., deSa, D. and Tabak, L.A. (1999)Structure-function analysis of the UDP-N-acetyl-D-galactosamine:poly-peptide N-acetylgalactosaminyltransferase. Essential residues lie in apredicted active site cleft resembling a lactose repressor fold. J. Biol.Chem., 274, 6797–6803.

27. Munro, S. and Freeman, M. (2000) The notch signalling regulator fringeacts in the Golgi apparatus and requires the glycosyltransferase signaturemotif DXD. Curr. Biol., 10, 813–820.

28. Taniguchi, N., Ekuni, A., Ko, J.H., Miyoshi, E., Ikeda, Y., Ihara, Y.,Nishikawa, A., Honke, K. and Takahashi, M. (2001) A glycomic approachto the identification and characterization of glycoprotein function in cellstransfected with glycosyltransferase genes. Proteomics, 1, 239–247.

29. Van den Steen, P.E., Opdenakker, G., Wormald, M.R., Dwek, R.A. andRudd, P.M. (2001) Matrix remodelling enzymes, the protease cascade andglycosylation. Biochim. Biophys. Acta, 1528, 61–73.

30. Jigami, Y. and Odani, T. (1999) Mannosylphosphate transfer to yeastmannan. Biochim. Biophys. Acta, 1426, 335–345.

31. Odani, T., Shimma, Y., Tanaka, A. and Jigami, Y. (1996) Cloning andanalysis of the MNN4 gene required for phosphorylation of N-linkedoligosaccharides in Saccharomyces cerevisiae. Glycobiology, 6, 805–810.

32. Odani, T., Shimma, Y., Wang, X.H. and Jigami, Y. (1997)Mannosylphosphate transfer to cell wall mannan is regulated by thetranscriptional level of the MNN4 gene in Saccharomyces cerevisiae. FEBSLett., 420, 186–190.

33. Henry, M.D. and Campbell, K.P. (1999) Dystroglycan inside and out. Curr.Opin. Cell Biol., 11, 602–607.

34. Winder, S.J. (2001) The complexities of dystroglycan. Trends Biochem.Sci., 26, 118–124.

35. Ervasti, J.M. and Campbell, K.P. (1993) A role for thedystrophin-glycoprotein complex as a transmembrane linker betweenlaminin and actin. J. Cell Biol., 122, 809–823.

36. Gee, S.H., Montanaro, F., Lindenbaum, M.H. and Carbonetto, S. (1994)Dystroglycan-alpha, a dystrophin-associated glycoprotein, is a functionalagrin receptor. Cell, 77, 675–686.

37. Sugiyama, J., Bowen, D.C. and Hall, Z.W. (1994) Dystroglycan binds nerveand muscle agrin. Neuron, 13, 103–115.

38. Hayashi, Y.K., Engvall, E., Arikawa-Hirasawa, E., Goto, K., Koga, R.,Nonaka, I., Sugita, H. and Arahata, K. (1993) Abnormal localization oflaminin subunits in muscular dystrophies. J. Neurol. Sci., 119, 53–64.

39. Matsubara, S., Mizuno, Y., Kitaguchi, T., Isozaki, E., Miyamoto, K. andHirai, S. (1999) Fukuyama-type congenital muscular dystrophy: closerelation between changes in the muscle basal lamina and plasma membrane[In Process Citation]. Neuromuscul. Disord., 9, 388–398.

40. Toda, T. and Kobayashi, K. (1999) Fukuyama-type congenital musculardystrophy: the first human disease to be caused by an ancientretrotransposal integration. J. Mol. Med., 77, 816–823.

41. Aoki, D., Lee, N., Yamaguchi, N., Dubois, C. and Fukuda, M.N. (1992)Golgi retention of a trans-Golgi membrane protein, galactosyltransferase,requires cysteine and histidine residues within the membrane-anchoringdomain. Proc. Natl Acad. Sci. USA, 89, 4319–4323.

42. Yamaguchi, N. and Fukuda, M.N. (1995) Golgi retention mechanism ofbeta-1,4-galactosyltransferase. Membrane-spanning domain-dependenthomodimerization and association with alpha- and beta-tubulins.J. Biol. Chem., 270, 12170–12176.

43. Herrmann, R., Straub, V., Blank, M., Kutzick, C., Franke, N., Jacob, E.N.,Lenard, H.G., Kroger, S. and Voit, T. (2000) Dissociation of thedystroglycan complex in caveolin-3-deficient limb girdle musculardystrophy. Hum. Mol. Genet., 9, 2335–2340.

44. Blake, D.J., Hawkes, R., Benson, M.A. and Beesley, P.W. (1999)Different dystrophin-like complexes are expressed in neurons and glia.J. Cell Biol., 147, 645–658.

Human Molecular Genetics, 2002, Vol. 11, No. 26 3331

Downloaded from https://academic.oup.com/hmg/article-abstract/11/26/3319/563489by gueston 11 February 2018