Elsevier

Neuroscience

Volume 220, 18 September 2012, Pages 47-61
Neuroscience

RPTPζ/phosphacan is abnormally glycosylated in a model of muscle–eye–brain disease lacking functional POMGnT1

https://doi.org/10.1016/j.neuroscience.2012.06.026Get rights and content

Abstract

Congenital muscular dystrophies (CMDs) with associated brain abnormalities are a group of disorders characterized by muscular dystrophy and brain and eye abnormalities that are frequently caused by mutations in known or putative glycotransferases involved in protein O-mannosyl glycosylation. Previous work identified α-dystroglycan as the major substrate for O-mannosylation and its altered glycosylation the major cause of these disorders. However, work from several labs indicated that other proteins in the brain are also O-mannosylated and therefore could contribute to CMD pathology in patients with mutations in the protein O-mannosylation pathway, however few of these proteins have been identified and fully characterized in CMDs. In this study we identify receptor protein tyrosine phosphatase ζ (RPTPζ) and its secreted variant, phosphacan, as another potentially important substrate for protein O-mannosylation in the brain. Using a mouse model of muscle–eye–brain disease lacking functional protein O-mannose β-1,2-N-acetylglucosaminyltransferase (POMGnT1), we show that RPTPζ/phosphacan is shifted to a lower molecular weight and distinct carbohydrate epitopes normally detected on the protein are either absent or substantially reduced, including Human Natural Killer-1 (HNK-1) reactivity. The spatial and temporal expression patterns of these O-mannosylated forms of RPTPζ/phosphacan and its hypoglycosylation and loss of HNK-1 glycan epitopes in POMGnT1 knockouts are suggestive of a role in the neural phenotypes observed in patients and animal models of CMDs.

Highlights

► A novel hypoglycosylated protein in POMGnT1 knockout brains, a model of CMDs. ► RPTPζ/phosphacan is a likely substrate for O-mannosylation by POMGnT1 in vivo. ► HNK-1 glycans on RPTPζ/phosphacan are diminished in POMGnT1 knockouts. ► Glycosylation of RPTPζ/phosphacan is unaffected in LARGE knockouts.

Introduction

Congenital muscular dystrophies (CMDs) with associated brain abnormalities are a group of devastating autosomal recessive disorders characterized by congenital muscular dystrophy, type II lissencephaly and eye anomalies (Dobyns et al., 1985, Haltia et al., 1997, Jimenez-Mallebrera et al., 2005, Reed, 2009). Abnormalities in protein O-mannosyl glycosylation, often arising from mutations in genes encoding functional or putative glycotransferase involved in O-mannosyl glycosylation, are causal to this group of disorders (Brockington et al., 2001a, Brockington et al., 2001b, Beltran-Valero de Bernabe et al., 2002, Schessl et al., 2006). Exemplary to this is muscle–eye–brain disease (MEB) which arises from mutations in the gene encoding the known glycotransferase protein O-mannose β-1,2-N-acetylglucosaminyltransferase 1 (POMGnT1) (Yoshida et al., 2001, Liu et al., 2006). POMGnT1 catalyzes the transfer of an N-acetylglucosamine saccharide on to O-mannose present on serine and threonine residues of glycoproteins (Yoshida et al., 2001).

The most well characterized protein substrate of O-mannosyl glycosylation is α-dystroglycan (α-DG), which forms a heterodimer with β-dystroglycan to function as a transmembrane cell-surface receptor for constituents of the extracellular matrix (ECM) present in the basal lamina of the developing and mature brain (Ervasti and Campbell, 1991, Ervasti and Campbell, 1993, Ibraghimov-Beskrovnaya et al., 1992, Montanaro et al., 1999). Proper protein O-mannosyl glycosylation of α-DG is essential to maintain its ligand-binding properties and the subsequent integrity of the basement membrane (Kano et al., 2002, Michele et al., 2002, Kim et al., 2004). Disruption of pial basement membrane integrity consequently leads to overmigration of cortical neurons and type II lissencephaly along with other CNS pathologies (Michele et al., 2002). A number of studies have demonstrated that altered glycosylation of α-DG accounts for most of the obvious neural abnormalities (Moore et al., 2002, Satz et al., 2010), however a number of pieces of evidence indicate that additional protein substrates for protein O-mannosyl glycosylation exist and may also contribute to the phenotypes observed in these disorders. Glycomic analysis revealed brains of animals lacking α-DG have similar amounts of proteins with O-mannose-initiated structures as wild-type animals (Stalnaker et al., 2011). Work also showed that up to 30% of all O-linked sugars in the brain are O-linked via mannose (Finne et al., 1979, Krusius et al., 1986, Chai et al., 1999, Kogelberg et al., 2001), a number that could not be accounted for by α-DG alone. Furthermore, O-mannosyl-linked carbohydrates have also been described on particular cell-adhesion molecules (Bleckmann et al., 2009). Therefore identifying and characterizing novel protein substrates with altered glycosylation in these CMDs may provide important insights into the molecular basis of their complex phenotypes. Perhaps most importantly, future avenues of therapeutic intervention may be revealed in the yet-unidentified molecular underpinnings of these disorders.

Our previous work demonstrated that the monoclonal antibody, Cat-315, likely detects a Human Natural Killer-1 (HNK-1) epitope present on receptor protein tyrosine phosphatase ζ (RPTPζ), also referred to as RPTPβ, in the developing brain with biochemical properties that correlated with O-linked mannose structures (Matthews et al., 2002, Dino et al., 2006) and in a neuroblastoma cell line (Abbott et al., 2008). RPTPζ along with its secreted variant, phosphacan (Maurel et al., 1994), are primarily expressed in the CNS (Shitara et al., 1994, Maeda et al., 1995) and have been implicated in several key developmental neural processes including proliferation (Ida et al., 2006, Soh et al., 2007), differentiation (Canoll et al., 1996, Ranjan and Hudson, 1996, Meng et al., 2000, Soh et al., 2007) cell-adhesion and migration (Abbott et al., 2008), axonal guidance and neurite outgrowth (Grumet et al., 1996, Sakurai et al., 1997, Hayashi et al., 2005), myelination (Harroch et al., 2000, Harroch et al., 2002) and higher order cognitive function (Niisato et al., 2005). The extracellular domains of RPTPζ/phosphacan bind a wide array of ligands important for normal CNS development (for review see (Peles et al., 1998)) including pleiotrophin (Maeda et al., 1996, Meng et al., 2000), midkine (Maeda et al., 1999), tenascin (Grumet et al., 1994, Milev et al., 1995), NCAM, Ng-CAM (Maurel et al., 1994, Milev et al., 1994) and contactin (Sakurai et al., 1997). Previous work has demonstrated that carbohydrate modifications on RPTPζ modulate receptor–ligand binding which can in-turn influence downstream signaling and cellular behaviors (Milev et al., 1995, Peles et al., 1998, Maeda et al., 2003, Abbott et al., 2008). Therefore, altered O-mannosylation of RPTPζ/phosphacan could potentially have deleterious effects on neural development.

The work presented herein demonstrates that RPTPζ/phosphacan is hypoglycosylated in POMGnT1 knockout mice, an animal model of MEB. These data suggest that RPTPζ/phosphacan is a novel and major protein substrate for protein O-mannosyl glycosylation by POMGnT1 in vivo. Furthermore we demonstrate that in the developing brain RPTPζ/phosphacan bears the majority of HNK-1 reactivity in the soluble fraction, and most of these glycans on RPTPζ/phosphacan are O-linked via mannose. Given the CNS-enriched expression and interactions of RPTPζ/phosphacan and the known importance of the HNK-1 carbohydrate in development, our data argue for an important role of altered O-mannosyl glycosylation on RPTPζ/phosphacan and a potential contribution to the complex neurological phenotypes associated with CMDs.

Section snippets

Animals

Protocols for animal usage were approved by the Institutional Animal Care and Use Committee of Upstate Medical University. POMGnT1 knockout mice were generated by Lexicon Genetics Incorporated (The Woodlands, TX, USA) (Liu et al., 2006). Large myodystrophy (Largemyd) mutant mice were acquired from Jackson Laboratories (Bar Harbor, ME, USA). For timed pregnant breeding, the morning of plug discovery was designated E0. POMGnT1 and Largemyd genotyping was carried out as previously published (

The immunoreactivity of monoclonal antibodies Cat-315 and 6B4 is dramatically reduced in the cortex of embryonic and early postnatal POMGnT1 knockout animals

Previous work definitively demonstrated the monoclonal antibody Cat-315 prominently detects an epitope on aggrecan in the adult brain (Matthews et al., 2002), but an epitope on RPTPζ/phosphacan in the immature brain (Dino et al., 2006). Further characterization showed that the Cat-315 antibody detects an O-linked carbohydrate epitope (Matthews et al., 2002, Dino et al., 2006), which we hypothesized was likely O-linked via mannose. To test this hypothesis, Cat-315 reactivity was evaluated in

Abnormal glycosylation of RPTPζ/phosphacan in models of CMDs with associated brain abnormalities

The congenital nature of CMD disorders, which stem from abnormalities in prenatal development provide a significant challenge to both diagnosing and treating affected individuals at the point when defects occur (Brasseur-Daudruy et al., 2011). Therefore, fully understanding the molecular pathomechanisms leading to the complex disease phenotypes of these disorders is imperative to future efforts in diagnostic testing and therapeutic intervention.

While this study primarily focused on a model

Conclusions

In summary, herein we reported that RPTPζ/phosphacan is hypoglycosylated in animal models of MEB lacking functional POMGnT1. These results suggest that RPTPζ/phosphacan is a significant substrate for protein O-mannosyl glycosylation in vivo, displaying the loss of several carbohydrate epitopes including HNK-1. Additionally, this work identifies the epitopes of several previously uncharacterized carbohydrate antibodies against RPTPζ/phosphacan as likely detecting O-mannosyl-linked glycans. While

Acknowledgements

The authors would like to thank Peng Zhang for animal genotyping and Wendi Burnette for technical assistance.

This work was funded by NIH/NINDS Grant #NS069660 to R.T.M and Grant #HD060458 and NS066582 to H.H.

Glossary

Receptor protein tyrosine phosphatase ζ (RPTPζ)/phosphacan
A chondroitin sulfate proteoglycan with CNS-enriched expression that is encoded by the PTPRZ1 gene. The full-length form of the protein functions as a receptor protein tyrosine phosphatase, while alternative splicing gives rise to a secreted form called phosphacan. While the full length- form of the receptor (RPTPζ) and secreted variant (phosphacan) are considered to be long-isoforms, short-isoforms of each variant have also been

References (84)

  • J. Garwood et al.

    Phosphacan short isoform, a novel non-proteoglycan variant of phosphacan/receptor protein tyrosine phosphatase-beta, interacts with neuronal receptors and promotes neurite outgrowth

    J Biol Chem

    (2003)
  • M. Grumet et al.

    Interactions with tenascin and differential effects on cell adhesion of neurocan and phosphacan, two major chondroitin sulfate proteoglycans of nervous tissue

    J Biol Chem

    (1994)
  • A. Haunso et al.

    Phosphacan immunoreactivity is associated with perineuronal nets around parvalbumin-expressing neurones

    Brain Res

    (1999)
  • N. Hayashi et al.

    Chondroitin sulfate proteoglycan phosphacan associates with parallel fibers and modulates axonal extension and fasciculation of cerebellar granule cells

    Mol Cell Neurosci

    (2005)
  • M. Ida et al.

    Identification and functions of chondroitin sulfate in the milieu of neural stem cells

    J Biol Chem

    (2006)
  • H. Kano et al.

    Deficiency of alpha-dystroglycan in muscle–eye–brain disease

    Biochem Biophys Res Commun

    (2002)
  • H. Kogelberg et al.

    NMR studies of mannitol-terminating oligosaccharides derived by reductive alkaline hydrolysis from brain glycoproteins

    Carbohydr Res

    (2001)
  • T. Krusius et al.

    Identification of an O-glycosidic mannose-linked sialylated tetrasaccharide and keratan sulfate oligosaccharides in the chondroitin sulfate proteoglycan of brain

    J Biol Chem

    (1986)
  • A. Kuga et al.

    Absence of post-phosphoryl modification in dystroglycanopathy mouse models and wild-type tissues expressing non-laminin binding form of alpha-dystroglycan

    J Biol Chem

    (2012)
  • J. Liu et al.

    A genetic model for muscle-eye-brain disease in mice lacking protein O-mannose 1,2-N-acetylglucosaminyltransferase (POMGnT1)

    Mech Dev

    (2006)
  • N. Maeda et al.

    Purification, characterization and developmental expression of a brain-specific chondroitin sulfate proteoglycan, 6B4 proteoglycan/phosphacan

    Neuroscience

    (1995)
  • N. Maeda et al.

    Multiple receptor-like protein tyrosine phosphatases in the form of chondroitin sulfate proteoglycan

    FEBS Lett

    (1994)
  • N. Maeda et al.

    Heterogeneity of the chondroitin sulfate portion of phosphacan/6B4 proteoglycan regulates its binding affinity for pleiotrophin/heparin binding growth-associated molecule

    J Biol Chem

    (2003)
  • N. Maeda et al.

    A receptor-like protein-tyrosine phosphatase PTPzeta/RPTPbeta binds a heparin-binding growth factor midkine. Involvement of arginine 78 of midkine in the high affinity binding to PTPzeta

    J Biol Chem

    (1999)
  • N. Maeda et al.

    6B4 proteoglycan/phosphacan, an extracellular variant of receptor-like protein-tyrosine phosphatase zeta/RPTPbeta, binds pleiotrophin/heparin-binding growth-associated molecule (HB-GAM)

    J Biol Chem

    (1996)
  • P. Milev et al.

    Complex-type asparagine-linked oligosaccharides on phosphacan and protein-tyrosine phosphatase-zeta/beta mediate their binding to neural cell adhesion molecules and tenascin

    J Biol Chem

    (1995)
  • I. Morita et al.

    HNK-1 glyco-epitope regulates the stability of the glutamate receptor subunit GluR2 on the neuronal cell surface

    J Biol Chem

    (2009)
  • I. Morita et al.

    HNK-1 (human natural killer-1) glyco-epitope is essential for normal spine morphogenesis in developing hippocampal neurons

    Neuroscience

    (2009)
  • F. Muntoni et al.

    The congenital muscular dystrophies in 2004: a century of exciting progress

    Neuromuscul Disord

    (2004)
  • E. Ong et al.

    Expression cloning of a human sulfotransferase that directs the synthesis of the HNK-1 glycan on the neural cell adhesion molecule and glycolipids

    J Biol Chem

    (1998)
  • E. Peles et al.

    Multi-ligand interactions with receptor-like protein tyrosine phosphatase beta: implications for intercellular signaling

    Trends Biochem Sci

    (1998)
  • M. Ranjan et al.

    Regulation of tyrosine phosphorylation and protein tyrosine phosphatases during oligodendrocyte differentiation

    Mol Cell Neurosci

    (1996)
  • J. Schessl et al.

    Congenital muscular dystrophies and the extracellular matrix

    Semin Pediatr Neurol

    (2006)
  • K. Shitara et al.

    Brain-specific receptor-type protein-tyrosine phosphatase RPTP beta is a chondroitin sulfate proteoglycan in vivo

    J Biol Chem

    (1994)
  • S.H. Stalnaker et al.

    Glycomic analyses of mouse models of congenital muscular dystrophy

    J Biol Chem

    (2011)
  • M.S. Viapiano et al.

    A novel membrane-associated glycovariant of BEHAB/brevican is up-regulated during rat brain development and in a rat model of invasive glioma

    J Biol Chem

    (2003)
  • H. Yagi et al.

    HNK-1 epitope-carrying tenascin-C spliced variant regulates the proliferation of mouse embryonic neural stem cells

    J Biol Chem

    (2010)
  • S. Yamamoto et al.

    Mice deficient in nervous system-specific carbohydrate epitope HNK-1 exhibit impaired synaptic plasticity and spatial learning

    J Biol Chem

    (2002)
  • A. Yoshida et al.

    Muscular dystrophy and neuronal migration disorder caused by mutations in a glycosyltransferase, POMGnT1

    Dev Cell

    (2001)
  • T. Yoshihara et al.

    Learning/memory impairment and reduced expression of the HNK-1 carbohydrate in beta4-galactosyltransferase-II-deficient mice

    J Biol Chem

    (2009)
  • C.T. Yuen et al.

    Brain contains HNK-1 immunoreactive O-glycans of the sulfoglucuronyl lactosamine series that terminate in 2-linked or 2,6-linked hexose (mannose)

    J Biol Chem

    (1997)
  • C. Bleckmann et al.

    O-glycosylation pattern of CD24 from mouse brain

    Biol Chem

    (2009)
  • Cited by (37)

    • Multiple distinct O-Mannosylation pathways in eukaryotes

      2019, Current Opinion in Structural Biology
    • Glycosylation with ribitol-phosphate in mammals: New insights into the O-mannosyl glycan

      2017, Biochimica et Biophysica Acta - General Subjects
      Citation Excerpt :

      However, how POMGNT1 is involved in the synthesis of post-phosphoryl modification on the core M3 structure remained unclear because the biosynthetic pathway of core M3 is independent of core M1 formation (Fig. 2). Human POMGNT1 is a type II membrane protein that has four domains: an N-terminal cytoplasmic domain [1–37], a transmembrane domain [38–59], a stem domain (60–299), and a catalytic domain (300–660) (Fig. 6A) [86]. The catalytic domain of POMGNT1 has homology with that of β1,2-N-acetylglucosaminyltransferase I, which catalyzes the formation of GlcNAcβ1-2Man in N-glycan [6].

    • The role of human natural killer-1 (HNK-1) carbohydrate in neuronal plasticity and disease

      2017, Biochimica et Biophysica Acta - General Subjects
      Citation Excerpt :

      The HNK-1 antigen appears to be highly immunogenic because several monoclonal antibodies, including L2, NC-1, M6749, VC1.1, Cat-315, and 6B4, show similar specificities to the HNK-1 antibody. Among them, the Cat-315 and 6B4 antibodies exhibit preferential specificity to the HNK-1 epitope expressed on O-glycans rather than on N-glycans [25–27] (Table 1). We recently found that autoantibodies of anti-MAG neuropathy patients also exhibit differential specificity to distinct HNK-1 epitopes depending on the carrier glycoproteins [28].

    View all citing articles on Scopus
    View full text