Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Miyabi Tanaka is active.

Publication


Featured researches published by Miyabi Tanaka.


American Journal of Human Genetics | 2008

Hyperglycosylation and Reduced GABA Currents of Mutated GABRB3 Polypeptide in Remitting Childhood Absence Epilepsy

Miyabi Tanaka; Richard W. Olsen; Marco T. Medina; Emily Schwartz; María Elisa Alonso; Reyna M. Durón; Ramon Castro-Ortega; Iris E. Martínez-Juárez; Ignacio Pascual-Castroviejo; Jesús Machado-Salas; Rene Silva; Julia N. Bailey; Dongsheng Bai; Adriana Ochoa; Aurelio Jara-Prado; Gregorio Pineda; Robert L. Macdonald; Antonio V. Delgado-Escueta

Childhood absence epilepsy (CAE) accounts for 10% to 12% of epilepsy in children under 16 years of age. We screened for mutations in the GABA(A) receptor (GABAR) beta 3 subunit gene (GABRB3) in 48 probands and families with remitting CAE. We found that four out of 48 families (8%) had mutations in GABRB3. One heterozygous missense mutation (P11S) in exon 1a segregated with four CAE-affected persons in one multiplex, two-generation Mexican family. P11S was also found in a singleton from Mexico. Another heterozygous missense mutation (S15F) was present in a singleton from Honduras. An exon 2 heterozygous missense mutation (G32R) was present in two CAE-affected persons and two persons affected with EEG-recorded spike and/or sharp wave in a two-generation Honduran family. All mutations were absent in 630 controls. We studied functions and possible pathogenicity by expressing mutations in HeLa cells with the use of Western blots and an in vitro translation and translocation system. Expression levels did not differ from those of controls, but all mutations showed hyperglycosylation in the in vitro translation and translocation system with canine microsomes. Functional analysis of human GABA(A) receptors (alpha 1 beta 3-v2 gamma 2S, alpha 1 beta 3-v2[P11S]gamma 2S, alpha 1 beta 3-v2[S15F]gamma 2S, and alpha 1 beta 3-v2[G32R]gamma 2S) transiently expressed in HEK293T cells with the use of rapid agonist application showed that each amino acid transversion in the beta 3-v2 subunit (P11S, S15F, and G32R) reduced GABA-evoked current density from whole cells. Mutated beta 3 subunit protein could thus cause absence seizures through a gain in glycosylation of mutated exon 1a and exon 2, affecting maturation and trafficking of GABAR from endoplasmic reticulum to cell surface and resulting in reduced GABA-evoked currents.


Neurology | 2008

Novel mutations in Myoclonin1/EFHC1 in sporadic and familial juvenile myoclonic epilepsy

Mayerlim Medina; Toshimitsu Suzuki; María Elisa Alonso; Reyna M. Durón; Iris E. Martínez-Juárez; Julia N. Bailey; Dongsheng Bai; Yushi Inoue; I. Yoshimura; Sunao Kaneko; M. C. Montoya; Adriana Ochoa; A. Jara Prado; Miyabi Tanaka; Jesús Machado-Salas; S. Fujimoto; M. Ito; S. Hamano; K. Sugita; Y. Ueda; Makiko Osawa; Hirokazu Oguni; Francisco Rubio-Donnadieu; Kazuhiro Yamakawa; Antonio V. Delgado-Escueta

Background: Juvenile myoclonic epilepsy (JME) accounts for 3 to 12% of all epilepsies. In 2004, the GENESS Consortium demonstrated four missense mutations in Myoclonin1/EFHC1 of chromosome 6p12.1 segregating in 20% of Hispanic families with JME. Objective: To examine what percentage of consecutive JME clinic cases have mutations in Myoclonin1/EFHC1. Methods: We screened 44 consecutive patients from Mexico and Honduras and 67 patients from Japan using heteroduplex analysis and direct sequencing. Results: We found five novel mutations in transcripts A and B of Myoclonin1/EFHC1. Two novel heterozygous missense mutations (c.755C>A and c.1523C>G) in transcript A occurred in both a singleton from Mexico and another singleton from Japan. A deletion/frameshift (C.789del.AV264fsx280) in transcript B was present in a mother and daughter from Mexico. A nonsense mutation (c.829C>T) in transcript B segregated in four clinically and seven epileptiform-EEG affected members of a large Honduran family. The same nonsense mutation (c.829C>T) occurred as a de novo mutation in a sporadic case. Finally, we found a three-base deletion (−364○%–362del.GAT) in the promoter region in a family from Japan. Conclusion: Nine percent of consecutive juvenile myoclonic epilepsy cases from Mexico and Honduras clinics and 3% of clinic patients from Japan carry mutations in Myoclonin1/EFCH1. These results represent the highest number and percentage of mutations found for a juvenile myoclonic epilepsy causing gene of any population group. GLOSSARY: CAE = childhood absence epilepsy; FS = febrile seizures in infancy/childhood; GM = grand mal tonic clonic seizure; JME = Juvenile myoclonic epilepsy; PSW = 3–6 Hz polyspike and slow wave complexes; SW = single spike and slow wave complex.


Epilepsia | 2010

GABRB3, epilepsy, and neurodevelopment

Miyabi Tanaka; Timothy M. DeLorey; Antonio V. Delgado-Escueta; Richard W. Olsen

GABRB3 is important to neurodevelopment, and appears to be influenced by non‐Mendelian and epigenetic mechanisms. GABRB3 abnormalities have been implicated in a variety of neurodevelopmental conditions presenting epilepsy phenotypes, including childhood absence epilepsy, Angelman syndrome, and autism. Gabrb3 disruption in mice also results in seizure phenotypes, ataxia, and sensory and learning disorders. For an expanded treatment of this topic see Jasper’s Basic Mechanisms of the Epilepsies, Fourth Edition (Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado‐Escueta AV, eds) published by Oxford University Press (available on the National Library of Medicine Bookshelf [NCBI] at http://www.ncbi.nlm.nih.gov/books).


Epilepsia | 2012

Effects on promoter activity of common SNPs in 5' region of GABRB3 exon 1A.

Miyabi Tanaka; Julia N. Bailey; Dongsheng Bai; Yumiko Ishikawa-Brush; Antonio V. Delgado-Escueta; Richard W. Olsen

Purpose:  The β3 subunit of the γ‐aminobutyric acid type A receptors (GABAA–Rs) is an essential component of GABAA–Rs in fetal, perinatal, and adult mammalian brain. Various transcripts of the β3 subunit gene (GABRB3) produce various proteins with different N‐termini. Rare variants in this N‐terminus (exon 1A and exon 2) of GABRB3 protein segregate in affected family members of two multigeneration‐multiplex families with remitting childhood absence epilepsy (rCAE), suggesting GABRB3 is a major Mendelian epilepsy gene for rare families with CAE. Therefore, the N‐terminus of GABRB3 could be important for GABRB3 regulation in development, and its alteration could produce rCAE. Herein we determine if single nucleotide polymorphisms (SNPs) within the 1,148‐bp region upstream from exon 1A influence the expression of GABRB3.


Epilepsia | 2009

DNA variants in coding region of EFHC1: SNPs do not associate with juvenile myoclonic epilepsy

Dongsheng Bai; Julia N. Bailey; Reyna M. Durón; María Elisa Alonso; Marco T. Medina; Iris E. Martínez-Juárez; Toshimitsu Suzuki; Jesús Machado-Salas; Ricardo Ramos-Ramírez; Miyabi Tanaka; Ramón H. Castro Ortega; Minerva López-Ruiz; Astrid Rasmussen; Adriana Ochoa; Aurelio Jara-Prado; Kazuhiro Yamakawa; Antonio V. Delgado-Escueta

Purpose:  Juvenile myoclonic epilepsy (JME) accounts for 3 to 12% of all epilepsies. In 2004, we identified a mutation‐harboring Mendelian gene that encodes a protein with one EF‐hand motif (EFHC1) in chromosome 6p12. We observed one doubly heterozygous and three heterozygous missense mutations in EFHC1 segregating as an autosomal dominant gene with 21 affected members of six Hispanic JME families from California and Mexico. In 2006, similar and three novel missense mutations were reported in sporadic and familial Caucasian JME from Italy and Austria. In this study, we asked if coding single nucleotide polymorphisms (SNPs) of EFHC1 also contribute as susceptibility alleles to JME with complex genetics.


Genetics in Medicine | 2017

EFHC1 variants in juvenile myoclonic epilepsy: reanalysis according to NHGRI and ACMG guidelines for assigning disease causality

Julia N. Bailey; Christopher Patterson; Laurence de Nijs; Reyna M. Durón; Viet-Huong Nguyen; Miyabi Tanaka; Marco T. Medina; Aurelio Jara-Prado; Iris E. Martínez-Juárez; Adriana Ochoa; Yolli Molina; Toshimitsu Suzuki; María Elisa Alonso; Jenny E. Wight; Yu-Chen Lin; Laura Maria de Figueiredo Ferreira Guilhoto; Elza Márcia Targas Yacubian; Jesús Machado-Salas; Andrea Daga; Kazuhiro Yamakawa; Thierry Grisar; Bernard Lakaye; Antonio V. Delgado-Escueta

Purpose:EFHC1 variants are the most common mutations in inherited myoclonic and grand mal clonic-tonic-clonic (CTC) convulsions of juvenile myoclonic epilepsy (JME). We reanalyzed 54 EFHC1 variants associated with epilepsy from 17 cohorts based on National Human Genome Research Institute (NHGRI) and American College of Medical Genetics and Genomics (ACMG) guidelines for interpretation of sequence variants.Methods:We calculated Bayesian LOD scores for variants in coinheritance, unconditional exact tests and odds ratios (OR) in case–control associations, allele frequencies in genome databases, and predictions for conservation/pathogenicity. We reviewed whether variants damage EFHC1 functions, whether efhc1−/− KO mice recapitulate CTC convulsions and “microdysgenesis” neuropathology, and whether supernumerary synaptic and dendritic phenotypes can be rescued in the fly model when EFHC1 is overexpressed. We rated strengths of evidence and applied ACMG combinatorial criteria for classifying variants.Results:Nine variants were classified as “pathogenic,” 14 as “likely pathogenic,” 9 as “benign,” and 2 as “likely benign.” Twenty variants of unknown significance had an insufficient number of ancestry-matched controls, but ORs exceeded 5 when compared with racial/ethnic-matched Exome Aggregation Consortium (ExAC) controls.Conclusions:NHGRI gene-level evidence and variant-level evidence establish EFHC1 as the first non–ion channel microtubule–associated protein whose mutations disturb R-type VDCC and TRPM2 calcium currents in overgrown synapses and dendrites within abnormally migrated dislocated neurons, thus explaining CTC convulsions and “microdysgenesis” neuropathology of JME.Genet Med 19 2, 144–156.


Experimental Neurology | 2012

Ontogeny of Lafora bodies and neurocytoskeleton changes in Laforin-deficient mice.

Jesús Machado-Salas; Maria Rosa Avila-Costa; Patricia Guevara; Jorge Guevara; Reyna M. Durón; Dongsheng Bai; Miyabi Tanaka; Kazuhiro Yamakawa; Antonio V. Delgado-Escueta

Lafora disease (LD) is an autosomal recessive, always fatal progressive myoclonus epilepsy with rapid cognitive and neurologic deterioration. One of the pathological hallmarks of LD is the presence of cytoplasmic PAS+polyglucosan inclusions called Lafora bodies (LBs). Current clinical and neuropathological views consider LBs to be the cause of neurological derangement of patients. A systematic study of the ontogeny and structural features of the LBs has not been done in the past. Therefore, we undertook a detailed microscopic analysis of the neuropile of a Laforin-deficient (epm2a-/-) mouse model. Wild type and epm2a-/- mice were sacrificed at different ages and their encephalon processed for light microscopy. Luxol-fast-blue, PAS, Bielschowski techniques, as well as immunocytochemistry (TUNEL, Caspase-3, Apaf-1, Cytochrome-C and Neurofilament L antibodies) were used. Young null mice (11 days old) showed necrotic neuronal death in the absence of LBs. Both cell death and LBs showed a progressive increment in size and number with age. Type I LBs emerged at two weeks of age and were distributed in somata and neurites. Type II LBs appeared around the second month of age and always showed a complex architecture and restricted to neuronal somata. Their number was considerably less than type I LBs. Bielschowski method showed neurofibrillary degeneration and senile-like plaques. These changes were more prominent in the hippocampus and ventral pons. Neurofibrillary tangles were already present in 11 days-old experimental animals, whereas senile-like plaques appeared around the third to fourth month of life. The encephalon of null mice was not uniformly affected: Diencephalic structures were spared, whereas cerebral cortex, basal ganglia, pons, hippocampus and cerebellum were notoriously affected. This uneven distribution was present even within the same structure, i.e., hippocampal sectors. Of special relevance, was the observation of the presence of immunoreactivity to neurofilament L on the external rim of type II LBs. Perhaps, type II LB is not the end point of a metabolic abnormality. Instead, we suggest that type II LB is a highly specialized structural and functional entity that emerges as a neuronal response to major carbohydrate metabolism impairment. Early necrotic cell death, neurocytoskeleton derangement, different structural and probably functional profiles for both forms of LBs, a potential relationship between the external rim of the LB type II and the cytoskeleton and an uneven distribution of these abnormalities indicate that LD is both a complex neurodegenerative disease and a glycogen metabolism disorder. Our findings are critical for future studies on disease mechanisms and therapies for LD. Interestingly, the neurodegenerative changes observed in this LD model can also be useful for understanding the process of dementia.


The New England Journal of Medicine | 2018

Variant Intestinal-Cell Kinase in Juvenile Myoclonic Epilepsy

Julia N. Bailey; Laurence de Nijs; Dongsheng Bai; Toshimitsu Suzuki; Hiroyuki Miyamoto; Miyabi Tanaka; Christopher Patterson; Yu-Chen Lin; Marco T. Medina; María Elisa Alonso; José M. Serratosa; Reyna M. Durón; Viet Nguyen; Jenny E. Wight; Iris E. Martínez-Juárez; Adriana Ochoa; Aurelio Jara-Prado; Laura Guilhoto; Yolly Molina; Elsa M. Yacubian; Minerva López-Ruiz; Yushi Inoue; Sunao Kaneko; Shinichi Hirose; Makiko Osawa; Hirokazu Oguni; Shinji Fujimoto; Thierry Grisar; John M. Stern; Kazuhiro Yamakawa

BACKGROUND In juvenile myoclonic epilepsy, data are limited on the genetic basis of networks promoting convulsions with diffuse polyspikes on electroencephalography (EEG) and the subtle microscopic brain dysplasia called microdysgenesis. METHODS Using Sanger sequencing, we sequenced the exomes of six members of a large family affected with juvenile myoclonic epilepsy and confirmed cosegregation in all 37 family members. We screened an additional 310 patients with this disorder for variants on DNA melting‐curve analysis and targeted real‐time DNA sequencing of the gene encoding intestinal‐cell kinase (ICK). We calculated Bayesian logarithm of the odds (LOD) scores for cosegregating variants, odds ratios in case–control associations, and allele frequencies in the Genome Aggregation Database. We performed functional tests of the effects of variants on mitosis, apoptosis, and radial neuroblast migration in vitro and conducted video‐EEG studies in mice lacking a copy of Ick. RESULTS A variant, K305T (c.914A→C), cosegregated with epilepsy or polyspikes on EEG in 12 members of the family affected with juvenile myoclonic epilepsy. We identified 21 pathogenic ICK variants in 22 of 310 additional patients (7%). Four strongly linked variants (K220E, K305T, A615T, and R632X) impaired mitosis, cell‐cycle exit, and radial neuroblast migration while promoting apoptosis. Tonic–clonic convulsions and polyspikes on EEG resembling seizures in human juvenile myoclonic epilepsy occurred more often in knockout heterozygous mice than in wild‐type mice (P=0.02) during light sleep with isoflurane anesthesia. CONCLUSIONS Our data provide evidence that heterozygous variants in ICK caused juvenile myoclonic epilepsy in 7% of the patients included in our analysis. Variant ICK affects cell processes that help explain microdysgenesis and polyspike networks observed on EEG in juvenile myoclonic epilepsy. (Funded by the National Institutes of Health and others.)


Frontiers in Molecular Neuroscience | 2018

IMPORTIN-8 MODULATES DIVISION OF APICAL PROGENITORS, DENDRITOGENESIS AND TANGENTIAL MIGRATION DURING DEVELOPMENT OF MOUSE CORTEX

Gerry Nganou; Carla Gomes Da Silva; Ivan Gladwyn-Ng; Dominique Engel; Bernard Coumans; Antonio V. Delgado-Escueta; Miyabi Tanaka; Laurent Nguyen; Thierry Grisar; Laurence de Nijs; Bernard Lakaye

The building of the brain is a multistep process that requires the coordinate expression of thousands of genes and an intense nucleocytoplasmic transport of RNA and proteins. This transport is mediated by karyopherins that comprise importins and exportins. Here, we investigated the role of the ß-importin, importin-8 (IPO8) during mouse cerebral corticogenesis as several of its cargoes have been shown to be essential during this process. First, we showed that Ipo8 mRNA is expressed in mouse brain at various embryonic ages with a clear signal in the sub-ventricular/ventricular zone (SVZ/VZ), the cerebral cortical plate (CP) and the ganglionic eminences. We found that acute knockdown of IPO8 in cortical progenitors reduced both their proliferation and cell cycle exit leading to the increase in apical progenitor pool without influencing the number of basal progenitors (BPs). Projection neurons ultimately reached their appropriate cerebral cortical layer, but their dendritogenesis was specifically affected, resulting in neurons with reduced dendrite complexity. IPO8 knockdown also slowed the migration of cortical interneurons. Together, our data demonstrate that IPO8 contribute to the coordination of several critical steps of cerebral cortex development. These results suggest that the impairment of IPO8 function might be associated with some diseases of neuronal migration defects.


Molecular Genetics & Genomic Medicine | 2016

Chromosome loci vary by juvenile myoclonic epilepsy subsyndromes: linkage and haplotype analysis applied to epilepsy and EEG 3.5-6.0 Hz polyspike waves.

Jenny E. Wight; Viet-Huong Nguyen; Marco T. Medina; Christopher Patterson; Reyna M. Durón; Yolly Molina; Yu-Chen Lin; Iris E. Martínez-Juárez; Adriana Ochoa; Aurelio Jara-Prado; Miyabi Tanaka; Dongsheng Bai; Sumaya Aftab; Julia N. Bailey; Antonio V. Delgado-Escueta

Juvenile myoclonic epilepsy (JME), the most common genetic epilepsy, remains enigmatic because it is considered one disease instead of several diseases. We ascertained three large multigenerational/multiplex JME pedigrees from Honduras with differing JME subsyndromes, including Childhood Absence Epilepsy evolving to JME (CAE/JME; pedigree 1), JME with adolescent onset pyknoleptic absence (JME/pA; pedigree 2), and classic JME (cJME; pedigree 3). All phenotypes were validated, including symptomatic persons with various epilepsies, asymptomatic persons with EEG 3.5–6.0 Hz polyspike waves, and asymptomatic persons with normal EEGs. Two‐point parametric linkage analyses were performed with 5185 single‐nucleotide polymorphisms on individual pedigrees and pooled pedigrees using four diagnostic models based on epilepsy/EEG diagnoses. Haplotype analyses of the entire genome were also performed for each individual. In pedigree 1, haplotyping identified a 34 cM region in 2q21.2–q31.1 cosegregating with all affected members, an area close to 2q14.3 identified by linkage (Zmax = 1.77; pedigree 1). In pedigree 2, linkage and haplotyping identified a 44 cM cosegregating region in 13q13.3–q31.2 (Zmax = 3.50 at 13q31.1; pooled pedigrees). In pedigree 3, haplotyping identified a 6 cM cosegregating region in 17q12. Possible cosegregation was also identified in 13q14.2 and 1q32 in pedigree 3, although this could not be definitively confirmed due to the presence of uninformative markers in key individuals. Differing chromosome regions identified in specific JME subsyndromes may contain separate JME disease‐causing genes, favoring the concept of JME as several distinct diseases. Whole‐exome sequencing will likely identify a CAE/JME gene in 2q21.2–2q31.1, a JME/pA gene in 13q13.3–q31.2, and a cJME gene in 17q12.

Collaboration


Dive into the Miyabi Tanaka's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dongsheng Bai

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco T. Medina

Universidad Nacional Autónoma de Honduras

View shared research outputs
Top Co-Authors

Avatar

Adriana Ochoa

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge