Myoclonin1/EFHC1 in cell division, neuroblast migration, synapse/dendrite formation in juvenile myoclonic epilepsy

This chapter presents recent works on Myocloni1/ EFHC1 a protein encoded by an epilepsy causing gene of juvenile myoclonic epilepsy (JME), one of the most frequent forms of idiopathic or genetic generalized epilepsies. Myoclonin 1/EFHC1 is a microtubule-associated protein involved in the regulation of cell division. In vitro, EFHC1 loss of function disrupted mitotic spindle organization, impaired M phase progression, induced microtubule bundling and increased apoptosis. EFHC1 impairment in the rat developing neocortex by ex vivo and in utero electroporation caused a marked disruption of radial migration. This effect was a result of cortical progenitors failing to exit the cell cycle. On the other hand, defects in the radial glia scaffold organization and in the locomotion of postmitotic neurons. Mutant analysis of Defhc1 lossand gain-of-function alleles in vivo in Drosophila revealed a number of neuronal defects, including abnormal synaptic development characterized by extensive satellite bouton formation, increased frequency of spontaneous neurotransmitter release, and aberrations in dendritic arbour morphogenesis. Thus, Myoclonin 1/ EFHC1 is a regulator of cell division and neuronal migration during cortical development synaptic bouton and dendritic morphogenesis. Disruption of these properties lead to JME, being now therefore considered as a developmental disease. Juvenile myoclonic epilepsy (JME) is the most frequent form of idiopathic/genetic generalized epilepsy. It accounts for 2–12 % of all epilepsies. JME symptoms of myoclonias and tonicclonic convulsions appear in adolescence in an otherwise normal person with normal neurological and cognitive functions (Delgado-Escueta and Bacsal, 1984)1. Five percent of these patients, five percent of their affected family members and eight percent of a 252 patient Corresponding author : Prof Thierry GRISAR, MD,PhD, University of Liege, GIGA Neuroscience Blvd de l’Hopital 1, 4000 Liege BELGIUM e-mail : tgrisar@ulg.ac.be. Japer's Basic M ecanism s of he Eppsies Japer's Basic M ecanism s of he Eppsies cohort followed for over 20 years only have seizures when triggered by external factors such as alcohol use, fatigue, menstruation and sleep deprivation. To understand the disease mechanisms underlying the stages of susceptibility and epileptogenesis in JME, many have tried to define its complex heritability and identify the genes corresponding to the fifteen chromosomal loci so far linked to the disease2 (see also chapter ...in this book). In 2004, Suzuki et al. identified several heterozygous missense mutations in a gene called EFHC1 3 in different unrelated families with JME probands. Since then, heterozygous non sense, deletion frame shifts and novel missense mutations have been identified in various populations of Italy, Austria and Chile 4–7 .Nine percent of sporadic JME cases detected in families consecutively seen in epilepsy clinics of Mexico and Honduras and 3 % of clinic patients from Japan carry mutations in EFHC1. This represents the highest number and percentage of mutations found for a juvenile myoclonic epilepsy causing gene of any population group 7. The gene encodes a 75-kDa protein with three DM10 domains of unknown function and a single EF-hand motif, a Ca2+-binding domain. The transcript is observed in many cell type of human tissue including brain (in particular in ependymal or periventricular cells) but also best expressed in dividing cells with highest levels in lung and testis 3. It was first proposed that EFHC1 was “pro-apoptotic” when overexpression in hippocampal neurons in vitro induced apoptotic cell death. This effect was significantly reduced by any of the five mutations associated with JME. Patch-clamp analysis of BHK (Baby Hamster Kidney) cells transfected with Cav2.3 VDCC (voltage-dependent calcium channel) and EFHC1 showed significantly increased R-type Ca2+ currents. So, the pro-apoptotic effect of EFHC1 was assigned to this enhancing effect on Ca2+ through Cav2.3 VDCC 3. In 2005, another research group pointed out that EFHC1 is orthologous to Rib72, an axonemal protein of Chlamydomonas reinhardtii. They demonstrated that EFHC1 is abundantly expressed in mouse tissues that have motile cilia or flagella, including the brain, and suggested that it plays a role in the intrinsic properties of these organelles 8. One of our laboratories previously reported that the subcellular distribution of EFHC1 in different cell lines varied during the cell cycle. In interphase cells, the protein is present in the cytoplasm and nucleus, except nucleoli and is particularly concentrated at the centrosome. During mitosis, EFHC1 is localized at spindle poles of the mitotic spindle and also at the midbody during cytokinesis 9. These results suggest that EFHC1 could play an important role during cell division and in particular during brain development since mRNA expression is higher at embryonic stages as compared to adult 10. More recently, we demonstrated that EFHC1 is a microtubule-associated protein (MAP) playing a key role in neuronal migration 11. In this chapter we review these putative roles of Myoclonin 1 / EFHC1 or during brain development and during adulthood. We posit an hypothesis that JME is a developmental disease involving neuronal migration and synaptic bouton and dendritic morphogenesis. EFHC1/MYOCLONIN1, A PROTEIN OF UNKNOWN FUNCTION Myoclonin 1 /EFHC1 gene is located on chromosome 6 (6p11–12) between markers D6S1960 and D6S11024, spans 72 kb and contains 11 exons. This gene encodes a protein of 640 amino acids. A domain search identified three tandemly repeated so called DM10 domains, a motif with unknown function. This protein also contains a single EF-hand, a well known Ca2+binding motif, from which it was named EFHC1 for EF-hand Containing 1 (Figure 1). This motif is located at the C-terminus between amino acid 578 and 606 and encoded by a nucleotide sequence present in exon 10. Page 2 Myoclonin1/EFHC1 in cell division, neuroblast migration, synapse/dendrite formation in juvenile myoclonic epilepsy Japer's Basic M ecanism s of he Eppsies Japer's Basic M ecanism s of he Eppsies Figure 1. Schematic representation of the EFHC1/Myoclonin1 gene, the long and short forms of the EFHC1 protein and the different mutations found co segregated with the JME phenotype The transcript undergo alternative splicing in exon 4, resulting in a C-terminally truncated protein, eliminating the EF-hand domain and two DM10 sequences. This last short form therefore only contains 278 amino acids, the first 240 being common with the entire molecule.

[1]  A. Delgado-Escueta,et al.  Distribution of EFHC1 or Myoclonin 1 in mouse neural structures , 2010, Epilepsy Research.

[2]  L. Nguyen,et al.  EFHC1 interacts with microtubules to regulate cell division and cortical development , 2009, Nature Neuroscience.

[3]  Cecilia Conde,et al.  Microtubule assembly, organization and dynamics in axons and dendrites , 2009, Nature Reviews Neuroscience.

[4]  T. Tsumoto,et al.  Efhc1 deficiency causes spontaneous myoclonus and increased seizure susceptibility. , 2009, Human molecular genetics.

[5]  Í. Lopes-Cendes,et al.  Expression Profile and Distribution of Efhc1 Gene Transcript During Rodent Brain Development , 2009, Journal of Molecular Neuroscience.

[6]  Í. Lopes-Cendes,et al.  Characterization of the C-terminal half of human juvenile myoclonic epilepsy protein EFHC1: dimer formation blocks Ca2+ and Mg2+ binding to its functional EF-hand. , 2008, Archives of biochemistry and biophysics.

[7]  M. T. Medina,et al.  Novel mutations in Myoclonin1/EFHC1 in sporadic and familial juvenile myoclonic epilepsy , 2008, Neurology.

[8]  K. Yamakawa,et al.  Sequential expression of Efhc1/myoclonin1 in choroid plexus and ependymal cell cilia. , 2008, Biochemical and biophysical research communications.

[9]  Ramanathan Arvind,et al.  Knot/Collier and Cut Control Different Aspects of Dendrite Cytoskeleton and Synergize to Define Final Arbor Shape , 2007, Neuron.

[10]  M. Segal,et al.  Doublecortin Supports the Development of Dendritic Arbors in Primary Hippocampal Neurons , 2007, Developmental Neuroscience.

[11]  T. Furuichi,et al.  Very-KIND, a KIND domain–containing RasGEF, controls dendrite growth by linking Ras small GTPases and MAP2 , 2007, The Journal of cell biology.

[12]  L. Tsai,et al.  Cep120 and TACCs Control Interkinetic Nuclear Migration and the Neural Progenitor Pool , 2007, Neuron.

[13]  P. Striano,et al.  Mutational Analysis of EFHC1 Gene in Italian Families with Juvenile Myoclonic Epilepsy , 2007, Epilepsia.

[14]  H. Vogel,et al.  Structures and metal-ion-binding properties of the Ca2+-binding helix-loop-helix EF-hand motifs. , 2007, The Biochemical journal.

[15]  A. Delgado-Escueta Advances in Genetics of Juvenile Myoclonic Epilepsies , 2007, Epilepsy currents.

[16]  K. Inokuchi,et al.  The microtubule destabilizer stathmin mediates the development of dendritic arbors in neuronal cells , 2007, Journal of Cell Science.

[17]  Xinnan Wang,et al.  Drosophila spichthyin inhibits BMP signaling and regulates synaptic growth and axonal microtubules , 2007, Nature Neuroscience.

[18]  S. M. Kolk,et al.  Doublecortin‐like, a microtubule‐associated protein expressed in radial glia, is crucial for neuronal precursor division and radial process stability , 2007, The European journal of neuroscience.

[19]  C Baumgartner,et al.  Idiopathic generalized epilepsy phenotypes associated with different EFHC1 mutations , 2006, Neurology.

[20]  P. Hedera,et al.  Mutations in the GABRA1 and EFHC1 genes are rare in familial juvenile myoclonic epilepsy , 2006, Epilepsy Research.

[21]  A. Delgado-Escueta,et al.  EFHC1, a protein mutated in juvenile myoclonic epilepsy, associates with the mitotic spindle through its N-terminus. , 2006, Experimental cell research.

[22]  S. Itohara,et al.  The Rho-GTPase cdc42 regulates neural progenitor fate at the apical surface , 2006, Nature Neuroscience.

[23]  A. Foidart,et al.  Some genetic and biochemical aspects of myoclonus , 2006, Neurophysiologie Clinique/Clinical Neurophysiology.

[24]  S. King Axonemal protofilament ribbons, DM10 domains, and the link to juvenile myoclonic epilepsy. , 2006, Cell motility and the cytoskeleton.

[25]  Hideyuki Okano,et al.  New Neurons Follow the Flow of Cerebrospinal Fluid in the Adult Brain , 2006, Science.

[26]  M. Feany,et al.  Disease-related phenotypes in a Drosophila model of hereditary spastic paraplegia are ameliorated by treatment with vinblastine. , 2005, The Journal of clinical investigation.

[27]  A. Kriegstein,et al.  LIS1 RNA interference blocks neural stem cell division, morphogenesis, and motility at multiple stages , 2005, The Journal of cell biology.

[28]  J. Yates,et al.  Proteomic Analysis of Isolated Chlamydomonas Centrioles Reveals Orthologs of Ciliary-Disease Genes , 2005, Current Biology.

[29]  Li-Huei Tsai,et al.  Nucleokinesis in Neuronal Migration , 2005, Neuron.

[30]  J. Delabie,et al.  Identification of a novel centrosome/microtubule-associated coiled-coil protein involved in cell-cycle progression and spindle organization , 2005, Oncogene.

[31]  M. Hirono,et al.  The mouse ortholog of EFHC1 implicated in juvenile myoclonic epilepsy is an axonemal protein widely conserved among organisms with motile cilia and flagella , 2005, FEBS letters.

[32]  K. Zinn,et al.  Drosophila Spastin Regulates Synaptic Microtubule Networks and Is Required for Normal Motor Function , 2004, PLoS biology.

[33]  M. T. Medina,et al.  Mutations in EFHC1 cause juvenile myoclonic epilepsy , 2004, Nature Genetics.

[34]  K. Broadie,et al.  The Hereditary Spastic Paraplegia Gene, spastin, Regulates Microtubule Stability to Modulate Synaptic Structure and Function , 2004, Current Biology.

[35]  A. Wynshaw-Boris,et al.  Lis1 and doublecortin function with dynein to mediate coupling of the nucleus to the centrosome in neuronal migration , 2004, The Journal of cell biology.

[36]  E. Drier,et al.  New Synaptic Bouton Formation Is Disrupted by Misregulation of Microtubule Stability in aPKC Mutants , 2004, Neuron.

[37]  L. Cassimeris,et al.  TOGp, the human homolog of XMAP215/Dis1, is required for centrosome integrity, spindle pole organization, and bipolar spindle assembly. , 2004, Molecular biology of the cell.

[38]  E. Parati,et al.  Neurosphere and neurosphere-forming cells: morphological and ultrastructural characterization , 2003, Brain Research.

[39]  M. Frotscher,et al.  Erratum: Reelin signaling directly affects radial glia morphology and biochemical maturation (Development vol. 130 (4597-4609)) , 2003 .

[40]  M. Frotscher,et al.  Reelin signaling directly affects radial glia morphology and biochemical maturation , 2003, Development.

[41]  Werner Waldhäusl,et al.  The ATP-dependent helicase RUVBL1/TIP49a associates with tubulin during mitosis. , 2003, Cell motility and the cytoskeleton.

[42]  R. Hart,et al.  Microtubules are critical for radial glial morphology: Possible regulation by MAPs and MARKs , 2003, Glia.

[43]  S. Sen,et al.  Dynamic association of a tumor amplified kinase, Aurora-A, with the centrosome and mitotic spindle. , 2003, Cell motility and the cytoskeleton.

[44]  K. Gull,et al.  γ-Tubulin Functions in the Nucleation of a Discrete Subset of Microtubules in the Eukaryotic Flagellum , 2003, Current Biology.

[45]  A. Wynshaw-Boris,et al.  Multiple Dose-Dependent Effects of Lis1 on Cerebral Cortical Development , 2003, The Journal of Neuroscience.

[46]  M. Hirono,et al.  Rib72, a Conserved Protein Associated with the Ribbon Compartment of Flagellar A-microtubules and Potentially Involved in the Linkage between Outer Doublet Microtubules* , 2003, The Journal of Biological Chemistry.

[47]  R. Patel-King,et al.  A Bipartite Ca2+-regulated Nucleoside-diphosphate Kinase System within theChlamydomonas Flagellum , 2002, The Journal of Biological Chemistry.

[48]  P. Hiesinger,et al.  Drosophila VAP-33A Directs Bouton Formation at Neuromuscular Junctions in a Dosage-Dependent Manner , 2002, Neuron.

[49]  W. Halfter,et al.  A Critical Function of the Pial Basement Membrane in Cortical Histogenesis , 2002, The Journal of Neuroscience.

[50]  C. Ball,et al.  Identification of genes periodically expressed in the human cell cycle and their expression in tumors. , 2002, Molecular biology of the cell.

[51]  L. Tsai,et al.  Life is a journey: a genetic look at neocortical development , 2002, Nature Reviews Genetics.

[52]  G. Phillips,et al.  Molecular mechanisms of calcium and magnesium binding to parvalbumin. , 2002, Biophysical journal.

[53]  Tim Hui-Ming Huang,et al.  Isolating human transcription factor targets by coupling chromatin immunoprecipitation and CpG island microarray analysis. , 2002, Genes & development.

[54]  Gerald M. Rubin,et al.  Drosophila Fragile X-Related Gene Regulates the MAP1B Homolog Futsch to Control Synaptic Structure and Function , 2001, Cell.

[55]  Jaime Grutzendler,et al.  Two modes of radial migration in early development of the cerebral cortex , 2001, Nature Neuroscience.

[56]  A. Lewit-Bentley,et al.  EF-hand calcium-binding proteins. , 2000, Current opinion in structural biology.

[57]  C. van Broeckhoven,et al.  Nucleoside diphosphate kinase beta (Nm23-R1/NDPKbeta) is associated with intermediate filaments and becomes upregulated upon cAMP-induced differentiation of rat C6 glioma. , 2000, Experimental cell research.

[58]  S. Gräslund,et al.  The spatial and temporal expression of Tekt1, a mouse tektin C homologue, during spermatogenesis suggest that it is involved in the development of the sperm tail basal body and axoneme. , 2000, European journal of cell biology.

[59]  G. Davis,et al.  Drosophila Futsch/22C10 Is a MAP1B-like Protein Required for Dendritic and Axonal Development , 2000, Neuron.

[60]  G. Davis,et al.  Drosophila Futsch Regulates Synaptic Microtubule Organization and Is Necessary for Synaptic Growth , 2000, Neuron.

[61]  T. Kaufman,et al.  Drosophila Centrosomin Protein is Required for Male Meiosis and Assembly of the Flagellar Axoneme , 1998, The Journal of cell biology.

[62]  E. A. Fajer,et al.  Centrosomal components immunologically related to tektins from ciliary and flagellar microtubules. , 1994, Journal of cell science.

[63]  A. Delgado-Escueta,et al.  Juvenile myoclonic epilepsy of Janz , 1984, Neurology.

[64]  R. Kretsinger,et al.  Carp muscle calcium-binding protein. II. Structure determination and general description. , 1973, The Journal of biological chemistry.

[65]  P. Rakić Mode of cell migration to the superficial layers of fetal monkey neocortex , 1972, The Journal of comparative neurology.

[66]  M. Ikura Calcium binding and conformational response in EF-hand proteins. , 1996, Trends in biochemical sciences.