Loss-of-function GJA12/Connexin47 mutations cause Pelizaeus–Merzbacher-like disease

https://doi.org/10.1016/j.mcn.2007.01.010Get rights and content

Abstract

Recessive mutations in GJA12/Cx47, the gene encoding the gap junction protein connexin47 (Cx47), cause Pelizaeus–Merzbacher-like disease (PMLD), which is characterized by severe CNS dysmyelination. Three missense PMLD mutations, P87S, Y269D and M283T, were expressed in communication-incompetent HeLa cells, and in each case the mutant proteins appeared to at least partially accumulate in the ER. Cells expressing each mutant did not pass Lucifer Yellow or neurobiotin in scrape loading assays, in contrast to robust transfer in cells expressing wild type Cx47. Dual whole-cell patch clamping of transfected Neuro2A cells demonstrated that none of the mutants formed functional channels, in contrast to wild type Cx47. Immunostaining sections of primate brains demonstrated that oligodendrocytes express Cx47, which is primarily localized to their cell bodies. Thus, the Cx47 mutants associated with PMLD likely disrupt the gap junction coupling between astrocytes and oligodendrocytes.

Introduction

Gap junction communication (GJC) facilitates the diffusion of ions and small molecules (less than 1000 Da) between two apposed cells, thereby enabling electrical coupling, metabolic cooperation, and spatial buffering of ions (Bruzzone et al., 1996). The molecular basis of GJC is the gap junction plaque, an aggregation of tens to thousands of individual channels. Two hemichannels (or connexons) on apposing cell membranes form the channel. Hemichannels are composed of six connexins – a family of highly conserved integral membrane proteins that are usually named according to their predicted molecular mass (Willecke et al., 2002). Individual hemichannels can be composed of one (homomeric) or more than one (heteromeric) type of connexin. Similarly, channels can be composed of hemichannels containing the same (homotypic) or different (heterotypic) connexins (Kumar and Gilula, 1996).

Anatomical and functional studies of the rodent CNS have demonstrated that astrocytes and oligodendrocytes are coupled by gap junctions (Massa and Mugnaini, 1982, Ransom and Kettenmann, 1990, Robinson et al., 1993, Rash et al., 1997), and that each cell type expresses different connexins (Dermietzel et al., 1989, Yamamoto et al., 1990, Scherer et al., 1995, Dermietzel et al., 1997, Ochalski et al., 1997, Nagy et al., 1999, Altevogt et al., 2002, Menichella et al., 2003, Odermatt et al., 2003, Kleopa et al., 2004). Astrocyte/astrocyte (A/A) coupling is likely mediated by Cx43/Cx43 and Cx30/Cx30 homotypic channels, and astrocyte/oligodendrocyte (A/O) coupling is most likely mediated by Cx43/Cx47 and Cx30/Cx32 heterotypic channels, with an uncertain contribution of Cx26/Cx32 channels (Rash et al., 2001, Nagy et al., 2003, Altevogt and Paul, 2004, Li et al., 2004). Cx32/Cx32 homotypic channels appear to form gap junctions between layers of CNS myelin sheaths (Kamasawa et al., 2005), as previously described in PNS myelin sheaths (Bergoffen et al., 1993, Scherer et al., 1995, Balice-Gordon et al., 1998, Meier et al., 2004). A/O heterotypic channels appear to be spatially restricted, with Cx43/Cx47 outnumbering Cx30/Cx32 channels at oligodendrocyte somata (Kleopa et al., 2004, Kamasawa et al., 2005). Oligodendrocytes also express Cx29, which does not appear to form gap junction plaques (Altevogt et al., 2002, Kleopa et al., 2004). Mutations in GJB1, the gene encoding Cx32, cause the X-linked form of Charcot–Marie–Tooth disease (CMT1X), an inherited demyelinating neuropathy (Bergoffen et al., 1993, Scherer and Kleopa, 2005). Clinical manifestations of CNS dysfunction in CMT1X are rare, associated with a few of the more than 300 different GJB1 mutations (Taylor et al., 2003), indicating that Cx30/Cx32 A/O coupling is not critical in humans.

Although CNS myelination in Gja12/cx47-null mice is minimally affected (Menichella et al., 2003, Odermatt et al., 2003), recessive GJA12/Cx47 mutations cause a devastating dysmyelinating disease in humans, called Pelizaeus–Merzbacher-like disease (PMLD; Uhlenberg et al., 2004, Bugiani et al., 2006). PMD itself is an X-linked dysmyelinating disorder caused by mutations in Proteolipid Protein 1 (PLP1), which encodes the main intrinsic membrane protein of CNS myelin (Garbern et al., 1999). PMLD patients are clinically similar to those with PMD, with nystagmus, spasticity, and ataxia, as well as widespread changes in CNS white matter by MRI imaging (Hudson et al., 2004), but do not have PLP1 mutations. The clinical phenotype and MRI findings in patients with recessive GJA12/Cx47 mutations (Uhlenberg et al., 2004, Bugiani et al., 2006) provide provocative evidence that Cx47-mediated GJC is essential for the proper functioning of oligodendrocytes. We show here that Cx47 is expressed by oligodendrocytes in the primate brain and that recessive missense mutations in GJA12/Cx47 cause loss-of-function. Thus, the Cx47 mutants associated with PMLD likely disrupt the A/O coupling that is mediated by Cx43/Cx47 heterotypic channels.

Section snippets

The sequence of human Cx47

In their report describing the GJA12/Cx47 mutations that cause PMLD, Uhlenberg et al. (2004) used the amino acid sequence derived from a human Cx47 cDNA clone (NM_020435) that included part of the 5’untranslated region (UTR). Because this 5′UTR contains a potential additional start codon (ATG) that is 9 nucleotides upstream of the ATG conventionally considered to be the connexin start codon, Uhlenberg et al. (2004) added 3 amino acids in their description of the mutations. This alternative ATG

PMLD mutations appear to cause simple loss-of-function

The PMLD patients described to date (Uhlenberg et al., 2004, Bugiani et al., 2006) have similar phenotypes – nystagmus noted in early infancy, and impaired motor development noted by 15 months. Different genotypes – homozygous P128frameshift, M283T, G233S, or L278frameshift mutations, compound heterozygotes for P87S/P327frameshift or Y269D/R237stop mutations – cause the same phenotype, including alleles that would be predicted to disrupt the protein (P128frameshift, R237stop, L278frameshift,

Mutant human Cx47 expression constructs

Using primers developed from a human Cx47 DNA sequence (GenBank accession number AF014643), we amplified the putative open reading frame of Cx47 by RT–PCR (SuperScript II, Invitrogen, Carlsbad, CA) from polyA RNA isolated from human brain, corpus callosum, or spinal cord (Clontech, Mountain View, CA), adding EcoRV and BamHI restriction sites at the 5′ and 3′ ends, respectively. The 5′ primer eliminated an unlikely alternative AUG that has been electronically translated in some sequences as the

Acknowledgments

We thank Jonathan Lee, Julia Beamesderfer, and Kate Hawk for technical assistance, Dr. Mike Koval for advice, Drs. Klaus Willecke and Bruce Nicholson for the HeLa cells, Dr. David Paul for the Cx47 antibody, Dr. Doug Rosene for the rhesus monkey tissue, the Neuropathology Core of the University of Pennsylvania Alzheimer Disease Core Center for human brain tissue, Dr. Jim Garbern for the ASPA antibody, Dr. Virginia Lee for the GFAP antibody, and Dr. Alex Gow for the CHOP antibody. Postmortem

References (91)

  • N. Kamasawa et al.

    Connexin-47 and connexin-32 in gap junctions of oligodendrocyte somata, myelin sheaths, paranodal loops and Schmidt–Lanterman incisures: implications for ionic homeostasis and potassium siphoning

    Neuroscience

    (2005)
  • J.G. Laing et al.

    Degradation of connexin43 gap junctions involves both the proteasome and the lysosome

    Exp. Cell Res.

    (1997)
  • X. Li et al.

    Connexin47, connexin29 and connexin32 co-expression in oligodendrocytes and Cx47 association with zonula occludens-1 (ZO-1) in mouse brain

    Neuroscience

    (2004)
  • Y. Ma et al.

    Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response

    J. Mol. Biol.

    (2002)
  • S. Melchionda et al.

    Functional characterization of a novel Cx26 (T55N) mutation associated to non-syndromic hearing loss

    Biochem. Biophys. Res. Commun.

    (2005)
  • P.J. Minogue et al.

    An aberrant sequence in a connexin46 mutant underlies congenital cataracts

    J. Biol. Chem.

    (2005)
  • J.I. Nagy et al.

    Connexin30 in rodent, cat and human brain: selective expression in gray matter astrocytes, co-localization with connexin43 at gap junctions and late developmental appearance

    Neuroscience

    (1999)
  • A. Oshima et al.

    Roles of Met-34, Cys-64, and Arg-75 in the assembly of human connexin 26. Implication for key amino acid residues for channel formation and function

    J. Biol. Chem.

    (2003)
  • W. Roscoe et al.

    Oculodentodigital dysplasia-causing connexin43 mutants are non-functional and exhibit dominant effects on wild-type connexin43

    J. Biol. Chem.

    (2005)
  • S.S. Scherer et al.

    X-linked Charcot–Marie–Tooth disease

  • C.R. Scriver et al.

    Monogenic traits are not simple: lessons from phenylketonuria

    Trends Genet.

    (1999)
  • A. Seki et al.

    Loss of electrical communication, but not plaque formation, after mutations in the cytoplasmic loop of connexin43

    Heart Rhy.

    (2004)
  • C.M. Southwood et al.

    The unfolded protein response modulates disease severity in Pelizaeus–Merzbacher disease

    Neuron

    (2002)
  • T. Thomas et al.

    Functional domain mapping and selective trans-dominant effects exhibited by Cx26 disease-causing mutations

    J. Biol. Chem.

    (2004)
  • J.E. Trosko et al.

    Gap junctions and the regulation of cellular functions of stem cells during development and differentiation

    Methods

    (2000)
  • B. Uhlenberg et al.

    Mutations in the gene encoding gap junction protein alpha 12 (connexin 46.6) cause Pelizaeus–Merzbacher-like disease

    Am. J. Hum. Genet.

    (2004)
  • M. Yeager et al.

    Structure of gap junction intercellular channels

    Curr. Opin. Struct. Biol.

    (1996)
  • S.W. Yum et al.

    Diverse trafficking abnormalities for connexin32 mutants causing CMTX

    Neurobiol. Dis.

    (2002)
  • C.K. Abrams et al.

    Pathogenesis of X-linked Charcot–Marie–Tooth disease: differential effects of two mutations in connexin 32

    J. Neurosci.

    (2003)
  • B.M. Altevogt et al.

    Four classes of intercellular channels between glial cells in the CNS

    J. Neurosci.

    (2004)
  • B.M. Altevogt et al.

    Cx29 is uniquely distributed within myelinating glial cells of the central and peripheral nervous systems

    J. Neurosci.

    (2002)
  • R.J. Balice-Gordon et al.

    Functional gap junctions in the Schwann cell myelin sheath

    J. Cell Biol.

    (1998)
  • J. Bergoffen et al.

    Connexin mutations in X-linked Charcot–Marie–Tooth disease

    Science

    (1993)
  • R. Bruzzone et al.

    Connections with connexins: the molecular basis of direct intercellular signaling

    Eur. J. Biochem.

    (1996)
  • M. Bugiani et al.

    GJA12 mutations in children with recessive hypomyelinating leukoencephalopathy

    Neurology

    (2006)
  • Y.H. Choung et al.

    Functional study of GJB2 in hereditary hearing loss

    Laryngoscope

    (2002)
  • J.E.A. Common et al.

    Cellular mechanisms of mutant connexins in skin disease and hearing loss

    Cell Commun. Adhes.

    (2003)
  • J. Das Sarma et al.

    Multimeric connexin interactions prior to the trans-Golgi network

    J. Cell Sci.

    (2001)
  • R. Dermietzel et al.

    Differential expression of three gap junction proteins in developing and mature brain tissues

    Proc. Natl. Acad. Sci. U.S.A.

    (1989)
  • R. Dermietzel et al.

    Oligodendrocytes express gap junction proteins connexin32 and connexin45

    Glia

    (1997)
  • S.M. Deschênes et al.

    Altered trafficking of mutant connexin32

    J. Neurosci.

    (1997)
  • W.L. Di et al.

    Defective trafficking and cell death is characteristic of skin disease-associated connexin 31 mutations

    Hum. Mol. Genet.

    (2002)
  • W.L. Di et al.

    Connexin interaction patterns in keratinocytes revealed morphologically and by FRET analysis

    J. Cell Sci.

    (2005)
  • C. Elfgang et al.

    Specific permeability and selective formation of gap junction channels in connexin-transfected HeLa cells

    J. Cell Biol.

    (1995)
  • G.M. Essenfelder et al.

    Connexin30 mutations responsible for hidrotic ectodermal dysplasia cause abnormal hemichannel activity

    Hum. Mol. Genet.

    (2004)
  • Cited by (97)

    • Glucose, glycolysis, and neurodegenerative disorders

      2023, Glycolysis: Tissue-Specific Metabolic Regulation in Physio-pathological Conditions
    • Activation of the unfolded protein response by Connexin47 mutations associated with Pelizaeus-Merzbacher-like disease

      2022, Molecular and Cellular Neuroscience
      Citation Excerpt :

      Our findings correlate with our previous studies conducted in HeLa and N2a cell lines where Cx47 mutant proteins associated with PMLD1 showed diffuse cytoplasmic staining compared to the WT punctuate staining (Orthmann-Murphy et al., 2007). Also, as previously reported, p.M283T showed both punctuate and ER localized staining while p.I33M showed punctuate staining similar to that of Cx47WT (Orthmann-Murphy et al., 2007; Orthmann-Murphy et al., 2009). Furthermore, the colocalization of an ER marker and Cx47 support the conclusion of our prior studies that suggested ER retention of Cx47 mutant proteins associated with PMLD1.

    View all citing articles on Scopus
    View full text