Advanced Medical Science(Cooperating field)Neurogenetics

Introduction

This division has started in September 2004 as a new section in the Center for Neurological Diseases and Cancer at the Nagoya University Graduate School of Medicine. Our major research interests include (1) physiology, pathomechanisms, and development of novel therapeutic options for neuromuscular signal transmission and other signal transmissions leading to muscle contractions, (2) molecular mechanisms and their aberrations of RNA processing especially of pre-mRNA splicing, (3) development of novel therapeutic options by exploiting the drug repositioning strategy for neuromuscular and skeletal disorders, (4) roles of intestinal microbiota on development of Parkinson’s disease, and (5) molecular mechanisms of divergent effects of molecular hydrogen on oxidative stress-mediated diseases.

Research Projects

1. Molecular mechanisms and development of therapeutic options for defective neuromuscular signal transmission

i) Congenital myasthenic syndromes

Congenital myasthenic syndromes (CMS) are heterogeneous disorders caused by germline mutations in genes expressed at the neuromuscular junction (Figure 1) (eLS DOI: 10.1002/9780470015902.a0024314, 2014). Mutations have been identified in 24 genes encoding acetylcholine receptor (AChR) subunits (CHRNA1, CHRNB1, CHRND, CHRNE and CHRNG), skeletal muscle sodium channel (SCN4A), signaling molecules driving clustering of AChR (AGRN, LRP4, MUSK and DOK7), synaptic structural proteins (COLQ, LAMB2 and COL13A1), postsynaptic structural proteins (RAPSN and PLEC), presynaptic molecules (CHAT and SYT2), glycosylation enzymes (GFPT1, DPAGT1, ALG2, ALG14 and GMPPB), and other less characterized molecules (PREPL and SCL25A1). CMS are recessive disorders, except for slow channel CMS caused by a missense mutation in one of AChR subunit genes and synaptotagmin 2 (SYT2)-CMS.
Onsets are mostly less than 2 years, but adult-onset is not rare, especially in slow-channel CMS and limb-girdle type CMS caused by glycosylation defects and by DOK7 mutations. Clinical features include fatigable muscle weakness, amyotrophy, and minor facial anomalies. Eye, facial and bulbar muscles are frequently affected, but sparing of these muscles is observed, especially in limb-girdle type CMS. We have first identified molecular defects in (i) choline acetyltransferase that resynthesizes acetylcholine from choline at the nerve terminal (Proc Natl Acad Sci U S A 98: 2017, 2001), (ii) collagen Q (ColQ) that anchors catalytic subunits of acetylcholinesterase to the synaptic basal lamina (Proc Natl Acad Sci U S A 95: 9654, 1998), (iii) AChR that opens cationic ion channel in response to acetylcholine released from the nerve terminal (Proc Natl Acad Sci U S A 92: 758, 1995, Neuron 17: 157, 1996), (iv) rapsyn that clusters AChR at the synaptic basal lamina (Am J Hum Genet 70: 875, 2002), (v) skeletal muscle voltage-gated sodium channel that senses membrane desensitization generated by AChR and opens sodium ion channel (Proc Natl Acad Sci U S A 100: 7377, 2003), and (vi) LRP4 that transmits a signal for clustering of AChR (Hum Mol Genet 23: 1856, 2014, JAMA Neurol 72: 889, 2015).
Only a single case of CMS has been genetically diagnosed in Japan before 2009. We started genetic diagnosis of CMS patients mostly using the next generation sequencing techniques and identified mutations in more than 20 patients. We proved that COLQ mutations impair binding of ColQ to MuSK (Figures 2 and 3) (Hum Mutat 34: 997, 2013). We also confirmed that introduction of the mutant ColQ into Colq-knockout mice using the protein-anchoring strategy, which is stated in the next section, does not ameliorate motor deficits (Figure 4). We additionally reported mutations in the AChR subunits with features of slow-channel syndrome and AChR deficiency (Figure 5) (Neuromuscul Disord 25: 60, 2015). Kinetic analysis of another slow-channel mutation in the AChR epsilon subunit gene revealed that a valine ring constituting a channel pore of AChR and comprised of five AChR subunits is critical for stabilizing AChR channel opening (Figure 6) (Hum Mutat 37: 1051, 2016). We are extensively looking for mutations in novel molecules and scrutinizing the underlying molecular pathomechanisms.

FIg01_CMS_Mutantions+Glcosylation.jpg Fig02_ColQ-MuSK_Scheme.jpg Fig03_COLQ_mutants.jpg
Figure 1. Molecules at the neuromuscular junction that are defective in congenital myasthenic syndromes. Figure 2. ColQ is anchored to the synaptic basal lamina by binding to MuSK and perlecan. Figure 3. Mutations in ColQ impair binding of ColQ to the neuromuscular junction of Colq-knockout mice.
Fig04_Protein_Anchoring_of_Mutant_ColQ.jpg Fig05_AChR_mutations_in_Japan.jpg Fig06_Valine_ring.jpg
Figure 4. Introduction of mutant D447H-COLQ using AAV8-mediated protein anchoring strategy fails to ameliorate motor deficits of Colq-knockout mice. Figure 5. Mutations (red symbols) identified in the acetylcholine receptor subunits in Japanese patients with congenital myasthenic syndrome. Single channel analysis revealed that the blue symbol is a normal polymorphism unique to Japanese population. Figure 6. Valine-to-alanine mutations at the valine ring of the channel pore of the acetylcholine receptor cause slow-channel syndrome.

ii) Protein-anchoring therapy

Gene therapy can essentially cure any kinds of cells modeling for human diseases. Treatment of human diseases with gene therapy, however, is hindered by unavailability of specific delivery of a transgene to the target organ. An extracellular matrix (ECM) protein carries proprietary binding domain(s), which target the protein to the property site(s). We exploited the unique feature of ECM proteins to ameliorate congenital defects of an ECM protein or to augment expression of an ECM protein.
There has been no rational therapy for endplate acetylcholinesterase (AChE) deficiency caused by genetic defects of COLQ encoding collagen Q. Asymmetric AChE species that are composed of AChE and collagen Q (Figure 7) are extracellular matrix molecules that are anchored to the synaptic basal lamina using the collagen domain and the C-terminal domain. We expected that introduction of AAV8-COLQ into skeletal muscles of Colq-knockout mice enables production of asymmetric AChE species, which is secreted from skeletal muscles and anchored to the synaptic basal lamina using its proprietary anchoring domains (Figure 8). We confirmed the feasibility of the protein-anchoring therapy with AAV8-COLQ and observed prominent effects in Colq-deficient mice (Figures 9, 10, and 11) (Mol Ther 20: 1384, 2012). We exploited this strategy to augment expression of biglycan in mdx mice modeling for Duchenne muscular dystrophy, and provided additional evidence that the protein-anchoring strategy can be applied to ECM molecules (Figure 12). (Hum Gene Ther in press).

Fig07_AChE_structure.jpg Fig08_Protein_anchoring_color.jpg Fig09_ColQ_movie.jpg
Figure 7. Six species of acetylcholinesterase (AChE) expressed in skeletal muscle. Figure 8. Protein-anchoring therapy for Colq-knockout mice. Figure 9. Protein-anchoring therapy improved movements of Colq-knockout mice (Fig09_ColQ_movie.mov).
Fig10_ColQ_exercise.jpg Fig11_ColQ_MEPP.jpg Fig12_NMJ_ECM.jpg
Figure 10. Protein-anchoring therapy normalized motor functions of Colq-knockout mice. Figure 11. Protein-anchoring therapy shortened MEPP decay time. Figure 12. Extracellular matrix proteins for which the protein-anchoring therapy is potentially effective.

iii) Identification of novel molecules essential for clustering of acetylcholine receptor (AChR)

In an effort to search for novel molecules that are essential for clustering of AChR at the neuromuscular junction (NMJ), we performed laser capture microdissection of mouse spinal motor neurons (SMNs) followed by gene expression profilings with microarray and RNA-seq. We found that a secreted activator of Wnt signaling, R-spondin 2 (Rspo2), is highly expressed in SMNs, and is enriched at the neuromuscular junction (Figure 13). Rspo2 induces MuSK phosphorylation and AChR clustering in cultured myotubes in an agrin-independent manner (Figure 14). Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) accumulated at the NMJ is associated with MuSK via LRP4, and serves as a receptor for Rspo2. In Rspo2-knockout mice, the number and density of AChRs at the NMJ are reduced, and the NMJ is ultrastructurally abnormal with widened synaptic clefts and sparse synaptic vesicles (Figure 15) (Sci Rep 6: 28512, 2016). We are also scrutinizing two other novel molecules that drive AChR clustering at the NMJ.

Fig13_LCM_of_mouse_spinal_cord.jpg Fig14_Rspo2_and_Lgr5.jpg Fig15_Rspo2_summary.jpg
Figure 13. Laser capture microdissection of the mouse spinal motor neurons (SMNs) reveals that Rspo2 is highly expressed in SMNs. Figure 14. Rspo2 induces clustering of acetylcholine receptor (AChR) in the absence of agrin. Knockdown of the Lgr5 receptor suppresses Rspo2-mediated clustering of AChR. Figure 15. Schematic of Rspo2-meidated clustering of the acetylcholine receptor.

iv) Dissection of molecular targets of anti-MuSK antibody

Myasthenia gravis is caused by anti-AChR, anti-MuSK, or anti-LRP4 antibodies. MuSK physiologically binds to LRP4 and ColQ, which constitutes the acetylcholinesterase/ColQ complex. We proved that anti-MuSK antibody blocks binding of ColQ to MuSK in vitro, and causes partial endplate acetylcholinesterase deficiency in mice that were passively transferred with patient’s anti-MuSK antibodies (Figure 16) (Neurology 77: 1819, 2011). We later proved that anti-MuSK antibody also blocks binding of MuSK to LRP4 in vitro in the presence of agrin. Passive transfer of anti-MuSK antibody to Colq-knockout mice similarly attenuated AChR clustering, indicating that AChR deficiency in anti-MuSK patients is likely due to blocking of LRP4, not of ColQ (Sci Rep 5: 13928, 2015). To our surprise, binding of ColQ to MuSK physiologically blocks MuSK-LRP4 interaction and suppresses agrin/LRP4/MuSK signaling. Quantitative analysis revealed that anti-MuSK antibody suppresses agrin/LRP4/MuSK signaling to a greater extent than the AChE/ColQ complex (Figure 17). This was why replacement of ColQ by anti-MuSK antibody attenuates agrin/LRP4/MuSK signaling. We are currently further dissecting mutual interactions between agrin, LRP4, MuSK, ColQ, biglycan, and anti-MuSK antibodies.

Fig16_Anti-MuSK_blocks_ColQ_and_LRP4.jpg Fig17_Anti-MuSK_Scheme.jpg
Figure 16. Anti-MuSK antibody blocks binding of ColQ to MuSK. Figure 17. Acetylcholinesterase (AChE)/ColQ complex blocks binding of MuSK to LRP4 and suppresses MuSK phosphorylation. Anti-MuSK antibody displaces AChE/ColQ and further blocks binding of MuSK to LRP4, and inhibits MuSK phosphorylation.

2. Physiological and pathological regulations of RNA metabolisms

i) Disclosure of hidden scenarios of splicing cis-elements, and development of tools to predict splicing consequences of mutations affecting splicing cis-elements

We humans exploit tissue-specific and developmental stage-specific alternative splicing to express more than 100,000 proteins from ~18,000 genes. More than 96% of human multi-exon genes are known to be alternatively spliced. Alternative splicing is achieved by splicing cis-elements on each gene and splicing trans-factors that are regulated in tissue-specific and developmental stage-specific manners (Figure 18). Classical splicing cis-elements include the branch point sequence, the polypyrimidine tract, and the 3’ and 5’ splice sites. These cis-elements are highly degenerative. The degeneracy makes prediction of splicing consequence of mutations that disrupt splicing cis-elements difficult and equivocal. We developed an algorithm that we named the SD Score to predict if a given mutation at the 5’ splice site affects pre-mRNA splicing or not. The SD Score algorithm is available at our web server (Figure 19) (Nucleic Acids Res 35: 5995, 2007). The human branch point consensus sequences had been simply derived from similarity to the highly conserved yeast branch point sequence of UACUAAC, which makes prediction of splicing consequences of branch point-disrupting mutations difficult. We have experimentally disclosed that the human branch point consensus sequence is yUnAy (Figure 20) (Nucleic Acids Res 36: 2257, 2008). In addition, a single nucleotide substitution at the first nucleotide of an exon causes aberrant splicing only when the polypyrimidine tract in the preceding intron is short (an AG-dependent 3’ splice site) (Figure 21) (Nucleic Acids Res 39: 4396, 2011). We have also developed IntSplice, which predicts splicing consequences of mutations from positions -50 to -3 from the 3’ end of an intron including the branch point sequence and the polypyrimidine tract (Figure 22) (J Hum Genet 2016). IntSplice is available on our web server. Only a few researchers in the world are interested in predicting the splicing effects of mutations. We are currently developing similar tools for mutations at the other sites.
Using similar techniques, we are developing a tool to predict pathogenicity of amino acid substitutions. Currently, the ROC curve of our tool is better than any other 24 previously reported evaluation tools.

Fig18_Spliceosome.jpg Fig19_SD_Score.jpg Fig20 Pictogram_and_WebLogo.jpg
Figure 18. Splicing cis-elements and trans-factors Figure 19. SD-Score algorithm predicts the splicing consequence of a mutation affecting the 5’ splice site with 97.1% sensitivity and 94.7% specificity Figure 20. Pictogram (A) and WebLogo (B) of human branch point consensus sequence
Fig21_First_Nucleotide.jpg Fig22_IntSplice.jpg
Figure 21. Mutations at the first nucleotide of an exon at AG-dependent 3’ splice sites with short polypyrimidine tracts cause aberrant splicing Figure 22. IntSplice web server predicts the splicing consequence of a mutation at intronic position -50 to -3 close to the 3’ end of an intron.

ii) Molecular mechanisms underlying disruption of splicing cis-elements in human diseases

We dissect molecular mechanisms underlying exonic and intronic splicing mutations that do not affect conventional splicing cis-elements. An intronic mutation in CHRNA1 encoding the AChR alpha subunit identified in a patient with CMS disrupts a splicing cis-element and decreases a binding affinity for hnRNP H (Figure 23) (Hum Mol Genet 17: 4022, 2008). We also proved that the polypyrimidine tract binding protein (PTB) also binds to a neighboring site (Hum Mol Genet 18: 1229, 2009). Another exonic mutation in another CMS patient disrupts binding of hnRNP L and de novo gains binding of hnRNP LL (Figure 24) (Sci Rep 3: 2931, 2013). Similarly, a mutation disrupting a splicing enhancing cis-element on exon 16 of COLQ encoding collagen Q, loses binding of SRSF1 and de novo gains binding of hnRNP H, which cause exclusive skipping of COLQ exon 16 (Figure 25) (Sci Rep 5: 13208, 2015). We are currently working on additional mutations disrupting other splicing cis-elements in congenital myasthenic syndromes and other diseases.

Fig23_CHRNA1_PTBP1_and_hnRNP_H.jpg Fig24_hnRNP_L.jpg Fig25_COLQ_SRSF1_hnRNP_H.jpg
Figure 23. CHRNA1 exon P3A is suppressed by hnRNP H and PTB. A mutation in a patient attenuates binding of hnRNP H and provokes exon recognition. Figure 24. Proline-rich region (PRR) of hnRNP L (L) binds to PTB and suppresses exon P3A of CHRNA1. A G-to-A mutation in a patient displaces hnRNP L and de novo gains binding of hnRNP LL (LL) that lacks PRR, and provokes exon recognition. Figure 25. Constitutive COLQ exon 16 is enhanced by SRSF1. A mutation disrupts binding of SRSF1, and de novo gains binding of hnRNP H, and causes exon skipping.

iii) Molecular mechanisms of alternative splicing of genes expressed at the neuromuscular junction (NMJ)

MUSK exon 10 is alternatively skipped in human, but not in mouse. Skipping of exon 10 disrupts a cysteine-rich region (Fz-CRD), which is essential for Wnt-mediated AChR clustering (Figure 26). Block-scanning mutagenesis, RNA-affinity purification, mass spectrometry, Western blotting, siRNA-mediated gene knockdown revealed that hnRNP C, YB-1 and hnRNP L bind to MUSK exon 10 and cause skipping of exon 10. Antibody-mediated in vitro protein depletion and scanning mutagenesis additionally revealed that hnRNP C promotes binding of YB-1 and hnRNP L to the immediate downstream sites and enhances exon skipping. Simultaneous tethering of two splicing trans-factors to the target confirmed the cooperative effect of YB-1 and hnRNP L on hnRNP C-mediated exon skipping. Search for a similar motif in the human genome revealed nine alternative exons that were individually or coordinately regulated by hnRNP C and YB-1 (Sci Rep 4: 6841, 2014).
Acetylcholinesterase (AChE), encoded by the ACHE gene, hydrolyzes the neurotransmitter acetylcholine to terminate synaptic transmission. Alternative splicing close to the 3' end generates three distinct isoforms of AChE-T, AChE-H and AChE-R (Figure 27). We found that hnRNP H binds to two specific G-runs in exon 5a of human ACHE and activates the distal alternative 3' splice site (ss) between exons 5a and 5b to generate AChET (Figure 28). Specific effect of hnRNP H was corroborated by siRNA-mediated knockdown and artificial tethering of hnRNP H. Furthermore, hnRNP H competes for binding of CstF64 to the overlapping binding sites in exon 5a, and suppresses the selection of a cryptic polyadenylation site (PAS), which additionally ensures transcription of the distal 3' ss required for the generation of AChE-T. Expression levels of hnRNP H were positively correlated with the proportions of the AChE-T isoform in three different cell lines. HnRNP H thus critically generates AChE-T by enhancing the distal 3' ss and by suppressing the cryptic PAS. Global analysis of CLIP-seq and RNA-seq also revealed that hnRNP H competitively regulates alternative 3' ss and alternative PAS in other genes. HnRNP H and CstF64 play pivotal roles in switching alternative splicing and alternative polyadenylation (Nucleic Acids Res, in press).
We are dissecting molecular mechanisms of more genes expressed at the neuromuscular junction.

Fig26_MUSK_exon_10.jpg Fig27_AChE_scheme.jpg Fig28_AChE_splicing_scheme.jpg
Figure 26. Fz-CRD of MuSK is encoded by exons 10 and 11. MUSK exon 10 is alternatively skipped. Binding of hnRNP C to exon 10 enhances binding of hnRNP L and YB-1 to its immediate downstream, and coordinately enhances skipping of exon 10. Figure 27. Schematic of alternative splicing of AChE isoforms and their membrane localizations. Figure 28. Schematic showing competition between hnRNP H-mediated activation of the distal 3’ ss generating AChET and CstF64-mediated activation of the cryptic polyadenylation site (UAUAAA) generating AChE-H and AChE-R. The cryptic (pA-1) and canonical (pA-2) PASs are marked by red and blue closed circles, respectively. HnRNP H-binding G-runs and CstF64-binding UG/Us are overlapping, and hnRNP H and CstF64 compete for binding to exon 5a.

iv) Extensive splicing analysis with exon array and RNA-seq

We use both Affymetrix exon array and RNA-seq to detect alternative and aberrant splicing events. In an effort to identify aberrant splicing events in myotonic dystrophy (DM1), we developed an analysis tool for the Affymetrix exon array. Extensive analysis of different parameters disclosed that Z-score of each exon best discriminates false- and true-positives (Figure 29) (J Hum Genet 57: 368, 2012).
Comparison of the exon array and RNA-seq currently leads to a tentative conclusion that exon array is better than RNA-seq for analyzing expression levels, which is likely because PCR is included in RNA-seq, but not in the exon array.

Fig29_Exon_array_tool.jpg
Figure 29. Deviation value (Z-score) best discriminates false- and true-positives of the Affymetrix exon array.

v) Elucidation of the roles of RNA-binding proteins by multi-OMICS analysis including CLIP-seq

MBNL1 and CUGBP1 are RNA-binding proteins that are misregulated in myotonic dystrophy (DM1). We determined RNA targets recognized by MBNL1 and CUGBP1 by CLIP-seq. We identified binding motifs (Figure 30) and position-specific splicing regulations of both proteins (Figure 31) (Sci Rep 2: 209, 2012). In addition, we found that both proteins preferentially bind to 3’ UTR and facilitate degradation of target transcripts (Figure 32). We additionally analyzed RNA targets of FUS by CLIP-seq, which is misregulated in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). We found that FUS binds to non-coding RNA arising from the antisense strands in the promoter regions and downregulates transcriptions of the target genes (Figure 33) (Sci Rep 2: 529, 2012). Multi-OMICS analyses using CLIP-seq, ChIP-seq, RNA-seq, CAGE-seq, and polyA-seq along with molecular dissections of individual FUS-regulated genes revealed that binding of FUS to nascent RNA stalls RNA polymerase II. If FUS binds immediately downstream to an alternative polyadenylation site, CPSF160 is recruited to the nascent RNA and polyadenylated short mRNA is generated, which also reduces the amount of polyadenylated long mRNA. If FUS binds immediately upstream to an alternative polyadenylation site, CPSF160 is not recruited to nascent RNA and the short nascent RNA transcript is degraded (Figure 34) (Genes Dev 29: 1045, 2015). We are developing a novel CLIP-seq technique to unveil yet unknown functions of RNA-binding proteins in a small scale.

Fig30_MBNL1_CUGBP1_motifs.jpg Fig31 MBNL1_CUGBP1_splicing.jpg Fig32_MBNL1_CUGBP1_3UTR.jpg
Figure 30. RNA-binding motifs of CUGBP1 and MBNL1. Figure 31. Position-specific splicing regulation by CUGBP1 and MBNL1. Figure 32. CUGBP1 and MBNL1 bind to 3’ UTRs and facilitate mRNA degradations.
Fig33_FUS_Promoter_antisense.jpg Fig34_FUS_polyA.jpg
Figure 33. FUS binds to non-coding RNA derived from an antisense strand in the promoter region. Figure 34. Position-specific binding of FUS to nascent RNA regulates mRNA length.

vi) siRNA-designing algorithm

RNA interference is commonly used as a modality for straightforward and robust suppression of the target gene. We applied our modeling techniques for designing siRNA, and developed an algorithm to design efficient siRNAs that we named iScore (Figures 35 and 36) (Nucleic Acids Res 35: e123, 2007). We have a web service program, iScore, which simultaneously calculates eight different siRNA-designing scores including iScore (Figure 37). Referring to the eight scores, we routinely make two siRNAs, and on average one of the two siRNAs suppresses the gene expression level down to more than 20%.

FIg35_iScore_ROC.jpg FIg36_iScore_Heat_ROC.jpg Fig37_iScore_web_service.jpg
Figure35.Sensitivity and specificity of the iScore siRNA-designing algorithm are comparable to those of three second-generation designing algorithms. Figure36.Heat-unstable siRNAs demonstrate an efficient ROC curve. Figure37. iScore designer web service program.

3. Drug repositioning strategy for orphan diseases

i) Repositioning of pre-approved drugs for neuromuscular disorders

Pre-approved drugs exhibit unpredicted beneficial effects on totally unrelated disorders, as observed in the anti-platelet effect of aspirin (NSAID); an anti-tremor effect of propranolol (adrenergic blocker); an anti-tumor effect of thalidomide (hypnotics); and anti-Parkinsonian effects of amantadine (anti-viral agent) and zonisamide (anti-epileptics) to name a few. We know optimal and maximal dosages, optimal routes and protocols of drug administrations, safety margins, and potential adverse effects of pre-approved drugs. With these advantages, information that we obtained in cultured cells and model animals can be readily applied to the clinical settings.
Slow channel syndrome is caused by prolonged opening episodes of the acetylcholine receptor. We have previously shown that anti-arrhythmic agent, quinidine (Ann Neurol 43: 480, 1998), and anti-depressant, fluoxetine (Neurology 60: 1710, 2003), ameliorate abnormally prolonged opening episodes of the acetylcholine receptor (Figure 38). Screening of neurite elongation of NSC34 cells revealed that an anti-epileptic and anti-Parkinsonian agent, zonisamide, enhances neurite elongation (Figure 39). We confirmed a dose-dependent effect using primary spinal motor neurons isolated from mice. Zonisamide markedly enhances axonal regeneration after an autograft operation of the sciatic nerve in mice (Figure 40) (PLoS One 10: e0142786, 2015). Zonisamide also exhibits effects on a mouse model of congenital myasthenic syndrome.
Myotonic dystrophy type 1 (DM1) is caused by abnormal expansion of CTG repeats in the 3’ untranslated region of the DMPK gene. Expanded CTG repeats are transcribed into RNA and make an aggregate with a splicing regulator, MBNL1, in the nucleus, which is called the nuclear foci. A nonsteroidal anti-inflammatory drug (NSAID), phenylbutazone (PBZ), upregulates the expression of MBNL1 in C2C12 mouse myoblasts as well as in a mouse model for DM1. In the DM1 mouse model, PBZ ameliorates aberrant splicing of Clcn1, Nfix, and Rpn2; increases expression of skeletal muscle chloride channel; decreases abnormal central nuclei of muscle fibers; and improves wheel-running activity. The effect of PBZ is attributed to two distinct mechanisms (Figure 41). First, PBZ suppresses methylation of an enhancer region in Mbnl1 intron 1, and enhances transcription of Mbnl1 mRNA. Second, PBZ attenuates binding of MBNL1 to abnormally expanded CUG repeats in cellulo and in vitro (Sci Rep 6: 25317, 2016). As PBZ is an old drug with unignorable adverse effects, we are currently looking for another pre-approved drug that exhibit similar effects.

Fig38_Quinidine_and_Fluoxetine.jpg Fig39_Zonisamide.jpg Fig40_Zonisamide_summary.jpg
Figure 38. Anti-arrhythmic agent, quinidine, and anti-depressant, fluoxetine, normalize abnormally prolonged ion channel openings in slow channel syndrome. Figure 39. Zonisamide induces neurite regeneration in the neurite scratch assay of mouse primary spinal motor neurons. Figure 40. Zonisamide is developed for epilepsy, and repositioned for Parkinson’s disease. Zonisamide can be repositioned again for peripheral nerve injuries, neuropathies, and neuromuscular disorders.
Fig41_Phenylbutazone.jpg
Figure 41. Phenylbutazone improves motor deficits in a mouse model for myotonic dystrophy type 1 by decreasing biding of MBNL1 to the abnormally expanded CUG repeats and by enhancing demethylation of Mbnl1 intron 1 followed by increased expression of Mbnl1.

ii) Repositioning of pre-approved drugs for skeletal disorders

In fibrodysplasia ossificans progressiva (FOP), we found that clinically applied Ca channel blockers, perhexiline and fendiline, are effective for model cells and model mice (Figure 42) (J Bone Miner Metab 31: 26, 2013). An open label trial with perhexiline in five FOP patients, however, showed no beneficial effects (Orphanet J Rare Dis 8: 163, 2013). We are looking for another pre-approved compound to treat FOP.
A proton pump inhibitor, lansoprazole, enhances nuclear accumulation of Runx2 and induces osteoblastogenesis (Figure 43). Systemic administration of lansoprazole to a rat femoral fracture model increased osteoblastogenesis. Dissection of signaling pathways revealed that lansoprazole activates a noncanonical bone morphogenic protein (BMP)-transforming growth factor-beta (TGF-beta) activated kinase-1 (TAK1)-p38 mitogen-activated protein kinase (MAPK) pathway. We found by in cellulo ubiquitination studies that lansoprazole enhances polyubiquitination of the TNF receptor-associated factor 6 (TRAF6) and by in vitro ubiquitination studies that the enhanced polyubiquitination of TRAF6 is attributed to the blocking of a deubiquitination enzyme, cylindromatosis (CYLD). Structural modeling and site-directed mutagenesis of CYLD demonstrated that lansoprazole tightly fits in a pocket of CYLD where the C-terminal tail of ubiquitin lies (Figure 44). Lansoprazole is a potential therapeutic agent for enhancing osteoblastic differentiation (Figure 45) (EBioMedicine 2: 2046, 2015).
Achondroplasia (ACH) is caused by gain-of-function mutations in FGFR3 encoding the fibroblast growth factor receptor 3. An anti-histamine and anti-emetic agent, meclozine, facilitates chondrocyte proliferation and mitigates loss of extracellular matrix in FGF2-treated rat chondrosarcoma (RCS) cells. Meclozine enhances proliferation of cultured cells modeling for ACH and embryonic tibial bone also modeling for ACH in explant culture (Figure 46) (PLoS One 8: e81569, 2013), as well as ACH model mice (Figure 47) (Endocrinology 156: 548, 2015). A Ca channel blocker, verapamil, induces expression of FRZB, a soluble antagonist of Wnt signaling in human osteoarthritis (OA) chondrocytes. Intraarticular injection of verapamil inhibits OA progression as well as nuclear localization of beta-catenin in a rat OA model (Figure 48) (PLoS One 9: e92699, 2014)
We are currently seeking for more pre-approved agents to treat skeletal disorders.

Fig42_FOP_uCT.jpg Fig43_Lansoprazole_osteogenesis.jpg Fig44_CYLD.jpg
Figure 42. Clinically applicable Ca channel blockers, perhexiline and fendiline, suppress heterotopic ossification in FOP model mice Figure 43. Lansoprazole increases matrix calcium deposition by Alizarin red staining in human bone marrow-derived mesenchymal cells that are induced to differentiate into osteoblasts. Figure 44. Simulated crystal structure of CYLD (PDB id: 2VHF) bound to ubiquitin (blue) (PDB id: 1NBF). The C-terminal tail of ubiquitin extends to the active site (red). Lansoprazole (green) fits into a pocket to cross the C-terminal tail of ubiquitin. Artificial mutations of R758 (not shown) and F766 (not shown) constituting the pocket to alanine have no effect on the enzymatic activity of CYLD, but nullify the effect of lansoprazole.
Fig45_Lansoprazole_summary.jpg Fig46_Meclzoine_explant_culture.jpg Fig47_Meclzoine_in_vivo.jpg
Figure 45. Proposed model of lansoprazole-induced activation of Runx2. Figure 46. Meclozine increases the longitudinal length of embryonic tibiae with or without FGF2 treatment in bone explant culture. Figure 47. Meclozine increases the longitudinal bone growth of Fgfr3ach mice.
Fig48_Verapamil.jpg
Figure 48. Intraarticular injection of verapamil ameliorates progression of osteoarthritis in a rat model.

4. Intestinal microbiota in Parkinson’s disease

Parkinson’s disease (PD) is characterized by abnormal aggregation of alpha-synuclein in substantia nigra and the other brain regions. It is currently established that abnormal accumulation of alpha-synuclein starts from the intestinal neural plexus and propagates into the dorsal nucleus of vagus nerve followed by locus ceruleus, and substantia nigra (Figure 49). In PD, intestinal permeability is increased and abnormal staining for E. coli and nitrotyrosine is observed in the intestinal wall. In an effort to search for the effects of intestinal microbiota in development of PD, we analyzed intestinal microbiota in 52 PD patients and 36 healthy cohabitants, and observed alterations in microbiota in PD. Modeling analysis revealed that Lactobacillus gasseri is elevated in advanced PD, and Clostridium coccoides is elevated in early PD (Figure 50). In addition, the number of hydrogen-producing bacteria is lower in PD patients compared to controls (Figure 51) (PLoS One 10: e0142164, 2015). We are currently recruiting more PD patients and healthy cohabitants, and analyzing intestinal microbiota using shotgun metagenome analysis and other biochemical markers.

Fig49_Brakk.jpg Fig50_Modeling.jpg Fig51_hydrogen_producing_bacteria.jpg
Figure 49. Braak’s observation showing that abnormal accumulation of alpha-synuclein ascends from dorsal nucleus of vagus up to substantia nigra. RBD, REM behavior disorder. Figure 50. Modeling analysis of gut microbiota in Parkinson’s disease reveals bacteria associated with progression of Parkinson’s disease and with severity of constipation. Figure 51. The number of hydrogen-producing bacteria is lower in Parkinson’s disease.

5. Elucidation of molecular mechanisms of the effects of molecular hydrogen on a plethora of diseases and disease models

Molecular hydrogen is effective for a wide range of disease models (Figure 52) and human diseases (Figure 53) (Oxid Med Cell Longev 2012: 353152, 2012). A total of 321 original articles have been published from 2007 to 2015 (Med Gas Res 5: 12, 2015). About three-quarters of the articles show the effects in mice and rats. The number of clinical trials is increasing every year. The effects have been reported in essentially all organs covering 31 disease categories that can be subdivided into 166 disease models, human diseases, treatment-associated pathologies, and pathophysiological conditions of plants with a predominance of oxidative stress-mediated diseases and inflammatory diseases. We reported the effects of hydrogen in a rat model of Parkinson’s disease (Figures 54 and 55) (Neurosci Lett 453: 81, 2009), patients with inflammatory and mitochondrial myopathies (Med Gas Res 1: 24, 2011), rat fetal hippocampal damage caused by in utero ischemia-reperfusion (Free Radic Biol Med 69: 324, 2014), pulmonary hypertension in rats (J Thorac Cardiovasc Surg 150: 645, 2015), mdx mice modeling for Duchenne muscular dystrophy (Redox Rep 1, 2016), perinatal brain injury caused by in utero prenatal inflammation (Free Radic Biol Med 91: 154, 2016), and bronchopulmonary dysplasia (BPD) in newborn rats (Pediatr Pulmonol 51: 928, 2016).
Specific extinction of hydroxyl radical and peroxynitrite was initially presented, but the radical-scavenging effect of hydrogen cannot be held solely accountable for its drastic effects. Molecular hydrogen suppresses FcepsilonRI-mediated signal transduction of mast cells (Biochem Biophys Res Commun 389: 651, 2009), and also inhibits lipopolysaccharide/interferon gamma-induced nitric oxide production through modulation of signal transduction in macrophages (Biochem Biophys Res Commun 411: 143, 2011). Extensive analyses of gene expression profiles also demonstrate that hydrogen modulates multiple signaling pathways (Mol Cell Biochem 403: 231, 2015). We also reported that hydrogen suppresses activated Wnt/beta-catenin signaling by enhancing the activity of the degradation complex of beta-catenin (Sci Rep 6: 31986, 2016). In addition, hydrogen water and intermittent hydrogen gas exposure, but not continuous hydrogen gas exposure, prevent 6-hydorxydopamine-induced Parkinson's disease in rats (Med Gas Res 2: 15, 2012). We are currently narrowing down to an exact target of hydrogen, which ignites activation and suppression of a wide range of signaling cascades.

Fig52_Hydrogen_animals.jpg Fig53_Hydogen_human.jpg
Figure 52. Representative disease models for which molecular hydrogen exhibits beneficial effects. Figure 53. Human diseases for which molecular hydrogen exhibits beneficial effects.

Fig54 Hydrogen_Ctr_Low.jpg

Fig55 Hydrogen_PreH_Low.jpg

Figure 54. A rat with hemi-Parkinsonism taking control water vigorously rotates clockwise after intraperitoneal injection of amphetamine (Fig54_Hydrogen_Ctr_Low.mov). Figure 55. A rat with hemi-Parkinsonism taking hydrogen-rich water does not rotate after intraperitoneal injection of amphetamine (Fig55_Hydrogen_PreH_Low.mov).

Faculty Members

FacultyPositionDepartment
Kinji Ohno Professor Neurogenetics
Akio Masuda Associate Professor Neurogenetics
Bisei Ohkawara Associate Professor Neurogenetics
Mikako Ito Designated Lecturer Neurogenetics
Jun-ichi Takeda Designated Assistant Professor Neurogenetics
Tomonari Hamaguchi Assistant Professor Neurogenetics

Bibliography

  • 2020
    1. Ozeki N, Yamawaki-Ogata A, Narita Y, Mii S, Ushida K, Ito M, Hirano S, Kurokawa R, Ohno K, Usui A. Hydrogen water alleviates obliterative airway disease in mice. Gen Thorac Cardiovasc Surg 2020, 68: 158-163.
    2. Ueyama J, Oda M, Hirayama M, Sugitate K, Sakui N, Hamada R, Ito M, Saito I, Ohno K. Freeze-drying enables homogeneous and stable sample preparation for determination of fecal short-chain fatty acids. Anal Biochem 2020, 589: 113508.
    3. Huang K, Masuda A, Chen G, Bushra S, Kamon M, Araki T, Kinoshita M, Ohkawara B, Ito M, Ohno K. Inhibition of cyclooxygenase-1 by nonsteroidal anti-inflammatory drugs demethylates MeR2 enhancer and promotes Mbnl1 transcription in myogenic cells. Sci Rep 2020, 10: 2558.
    4. Nakazawa Y, Hara Y, Oka Y, Komine O, van den Heuvel D, Guo C, Daigaku Y, Isono M, He Y, Shimada M, Kato K, Jia N, Hashimoto S, Kotani Y, Miyoshi Y, Tanaka M, Sobue A, Mitsutake N, Suganami T, Masuda A, Ohno K, Nakada S, Mashimo T, Yamanaka K, Luijsterburg MS, Ogi T. Ubiquitination of DNA Damage-Stalled RNAPII Promotes Transcription-Coupled Repair. Cell 2020, 180: 1228-1244 e24.
    5. Masuda A, Kawachi T, Takeda JI, Ohkawara B, Ito M, Ohno K. tRIP-seq reveals repression of premature polyadenylation by co-transcriptional FUS-U1 snRNP assembly. EMBO Rep 2020, 21: e49890.
    6. Kusano T, Nakatani M, Ishiguro N, Ohno K, Yamamoto N, Morita M, Yamada H, Uezumi A, Tsuchida K. Desloratadine inhibits heterotopic ossification by suppression of BMP2-Smad1/5/8 signaling. J Orthop Res in press.
    7. Ohkawara B, Shen X-M, Selcen D, Nazim M, Bril V, Tarnopolsky MA, Brady L, Fukami S, Amato AA, Yis U, Ohno K, Engel AG. Agrin Myasthenia: Distinct effects of variants in different domains on clustering of acetylcholine receptors. JCI Insight in press.
    8. Nishiwaki H, Ito M, Ishida T, Hamaguchi T, Maeda T, Kashihara K, Tsuboi Y, Ueyama J, Shimamura T, Mori H, Kurokawa K, Katsuno M, Hirayama M, Ohno K. Meta-Analysis of Gut Dysbiosis in Parkinson’s Disease. Mov Disord in press.
    9. Takeda J, Nanatsue K, Yamagishi R, Ito M, Haga N, Hirata H, Ohno K. InMeRF: Prediction of pathogenicity of missense variants by individual modeling for each amino acid substitution. NAR Genomics and Bioinformatics in press.
    10. Ohkawara B*, Kobayakawa A*, Kanbara S, Hattori T, Kubota S, Ito M, Masuda A, Takigawa M, Lyons KM, Ishiguro N, Ohno K. CTGF/CCN2 facilitates LRP4-mediated formation of the embryonic neuromuscular junction. EMBO Rep in press. *Equal contribution
  • 2019
    1. Okura T, Ohkawara B, Takegami Y, Ito M, Masuda A, Seki T, Ishiguro N, Ohno K. Mianserin suppresses R-spondin 2-induced activation of Wnt/β-catenin signaling in chondrocytes and prevents cartilage degradation in a rat model of osteoarthritis. Sci Rep 2019, 9: 2808.
    2. Abe K, Hirayama M, Ohno K, Shimamura T. ENIGMA: an enterotype-like unigram mixture model for microbial association analysis. BMC Genomics 2019, 20: 191.
    3. Tsuda T, Nonome T, Goto S, Takeda J, Tsunoda M, Hirayama M, Ohno K. Application of Skin Gas GC/MS Analysis for Prediction of the Severity Scale of Parkinson’s Disease. Chromatography 2019, 40: 149-155.
    4. Kataoka N, Maeda A, Ohno K. RNA Diseases in Humans – From Fundamental Research to Therapeutic Applications. Front Mol Biosci, 2019, 6:53 (査読有)
  • 2018
    1. Yu Y, Lin Y, Takasaki Y, Wang C, Kimura H, Xing J, Ishizuka K, Toyama M, Kushima I, Mori D, Arioka Y, Uno Y, Shiino T, Nakamura Y, Okada T, Morikawa M, Ikeda M, Iwata N, Okahisa Y, Takaki M, Sakamoto S, Someya T, Egawa J, Usami M, Kodaira M, Yoshimi A, Oya-Ito T, Aleksic B, Ohno K, Ozaki N. Rare loss of function mutations in N-methyl-D-aspartate glutamate receptors and their contributions to schizophrenia susceptibility. Transl Psychiatry 2018, 8: 12.
    2. Kurahashi H, Azuma Y, Masuda A, Okuno T, Nakahara E, Imamura T, Saitoh M, Mizuguchi M, Shimizu T, Ohno K, Okumura A. MYRF is associated with encephalopathy with reversible myelin vacuolization. Ann Neurol 2018, 83: 98-106.
    3. Ito K*, Ohkawara B*, Yagi H, Nakashima H, Tsushima M, Ota K, Konishi H, Masuda A, Imagama S, Kiyama H, Ishiguro N, Ohno K. Lack of Fgf18 causes abnormal clustering of motor nerve terminals at the neuromuscular junction with reduced acetylcholine receptor clusters. Sci Rep 2018, 8: 434. *Equal contribution.
    4. Nishiwaki H, Ito M, Negishi S, Sobue S, Ichihara M, Ohno K. Molecular hydrogen upregulates heat shock response and collagen biosynthesis, and downregulates cell cycles: meta-analyses of gene expression profiles. Free Radic Res 2018, 52: 434-445.
    5. Takeuchi A, Iida K, Tsubota T, Hosokawa M, Denawa M, Brown JB, Ninomiya K, Ito M, Kimura H, Abe T, Kiyonari H, Ohno K, Hagiwara M. Loss of Sfpq causes long-gene transcriptopathy in the brain. Cell Rep 2018, 23: 1326-1341.
    6. Li J, Ito M, Ohkawara B, Masuda A, Ohno K. Differential effects of spinal motor neuron-derived and skeletal muscle-derived Rspo2 on acetylcholine receptor clustering at the neuromuscular junction. Sci Rep 2018, 8: 13577.
    7. Abe K, Hirayama M, Ohno K, Shimamura T. A latent allocation model for the analysis of microbial composition and disease. BMC Bioinformatics 2018, 19: 519.
    8. Hirayama M, Ito M, Minato T, Yoritaka A, LeBaron TW, Ohno K. Inhalation of hydrogen gas elevates urinary 8-hydroxy-2'-deoxyguanine in Parkinson's disease. Med Gas Res 2018, 8: 144-149.
    9. Suzuki A, Ito M, Hamaguchi T, Mori H, Takeda Y, Baba R, Watanabe T, Kurokawa K, Asakawa S, Hirayama M, Ohno K. Quantification of hydrogen production by intestinal bacteria that are specifically dysregulated in Parkinson's disease. PLoS One 2018, 13: e0208313.
    10. Yoritaka A, Ohtsuka C, Maeda T, Hirayama M, Abe T, Watanabe H, Saiki H, Oyama G, Fukae J, Shimo Y, Hatano T, Kawajiri S, Okuma Y, Machida Y, Miwa H, Suzuki C, Kazama A, Tomiyama M, Kihara T, Hirasawa M, Shimura H, Oda E, Ito M, Ohno K, Hattori N. Randomized, double-blind, multicenter trial of hydrogen water for Parkinson's disease. Mov Disord 2018, 33: 1505-1507.
    11. Ohno K, Takeda JI, Masuda A. Rules and tools to predict the splicing effects of exonic and intronic mutations. Wiley Interdiscip Rev RNA 2018, 9: e1451. (査読有)
    12. Ito M, Ohno K. Protein-anchoring therapy to target extracellular matrix proteins to their physiological destinations. Matrix Biol, 2018, 68-69: 628-636. (査読有)
  • 2017
    1. Nazim M, Masuda A, Rahman MA, Nasrin F, Takeda JI, Ohe K, Ohkawara B, Ito M, Ohno K. Competitive regulation of alternative splicing and alternative polyadenylation by hnRNP H and CstF64 determines acetylcholinesterase isoforms. Nucleic Acids Res 2017, 45: 1455-1468.
    2. Hasegawa S, Ito M, Fukami M, Hashimoto M, Hirayama M, Ohno K. Molecular hydrogen alleviates motor deficits and muscle degeneration in mdx mice. Redox Rep 2017, 22: 26-34.
    3. Matsushita M, Mishima K, Esaki R, Ishiguro N, Ohno K, Kitoh H. Maternal administration of meclozine for the treatment of foramen magnum stenosis in transgenic mice with achondroplasia. J Neurosurg Pediatr 2017, 19: 91-95.
    4. Ito M*, Ehara Y*, Li J, Inada K, Ohno K. Protein-anchoring therapy of biglycan for mdx mouse model of Duchenne muscular dystrophy. Hum Gene Ther 2017, 28: 428-436. *Equal contribution.
    5. Takeda JI, Masuda A, Ohno K. Six GU-rich (6GUR) FUS-binding motifs detected by normalization of CLIP-seq by Nascent-seq. Gene 2017, 518: 57-64.
    6. Kishimoto Y, Ohkawara B, Sakai T, Ito M, Masuda A, Ishiguro N, Shukunami C, Docheva D, Ohno K. Wnt/β-catenin signaling suppresses expressions of Scx, Mkx, and Tnmd in tendon-derived cells. PLoS One, 2017, 12: e0182051.
    7. Ahsan KB, Masuda A, Rahman MA, Takeda J, Nazim M, Ohkawara B, Ito M, Ohno K. SRSF1 suppresses selection of intron-distal 5’ splice site of DOK7 intron 4 to generate functional full-length Dok-7 protein. Sci Rep 2017, 7: 10446.
    8. Tabeta K, Du X, Arimatsu K, Yokoji M, Takahashi N, Amizuka N, Hasegawa T, Crozat K, Maekawa T, Miyauchi S, Matsuda Y, Ida T, Kaku M, Hoebe K, Ohno K, Yoshie H, Yamazaki K, Moresco EM, Beutler B. An ENU-induced splice site mutation of mouse Col1a1 causing recessive osteogenesis imperfecta and revealing a novel splicing rescue. Sci Rep 2017, 7: 11717.
    9. Miyamoto K, Ohkawra B, Ito M, Masuda A, Hirakawa A, Sakai T, Hiraiwa H, Hamada T, Ishiguro N, Ohno K. Fluoxetine ameliorates cartilage degradation in osteoarthritis by inhibiting Wnt/β-catenin signaling. PLoS One 2017, 12: e0184388.
    10. Minato T, Maeda T, Fujisawa Y, Tsuji H, Nomoto K, Ohno K, Hirayama M. Progression of Parkinson's disease is associated with gut dysbiosis: Two-year follow-up study. PLoS One 2017, 12: e0187307.
    11. Gao KP, Ren YC, Wang JJ, Liu ZC, Li JN, Li LL, Wang BY, Li H, Wang YX, Cao YK, Ohno K, Zhai RH, Liang Z. Interactions between genetic polymorphisms of glucose metabolizing genes and smoking and alcohol consumption in the risk of type 2 diabetes mellitus. Appl Physiol Nutr Med 2017, 42: 1316-1321.
    12. Kasai T, Nakatani M, Ishiguro N, Ohno K, Yamamoto N, Morita M, Yamada H, Tsuchida K, Uezumi A. Promethazine hydrochloride inhibits ectopic fat cell formation in skeletal muscle. Am J Pathol 2017, 187: 2627-2634.
    13. Osawa Y, Matsushita M, Hasegawa S, Esaki R, Fujio M, Ohkawara B, Ishiguro N, Ohno K, Kitoh H. Activated FGFR3 promotes bone formation via accelerating endochondral ossification in mouse model of distraction osteogenesis. Bone 2017, 105: 42-49.
    14. Ohno K, Rahman MA, Nazim M, Nasrin F, Lin Y, Takeda JI, Masuda A. Splicing regulation and dysregulation of cholinergic genes expressed at the neuromuscular junction. J Neurochem 2017, 142 Suppl 2: 64-72. (査読有)
    15. Ohno K, Ohkawara B, Ito M. Agrin-LRP4-MuSK signaling as a therapeutic target for myasthenia gravis and other neuromuscular disorders. Expert Opin Ther Targets 2017, 21: 949-958.(査読有)
  • 2016
    1. Yagi H, Ohkawara B, Nakashima H, Ito K, Tsushima M, Ishii H, Noto K, Ohta K, Masuda M, Imagama S, Ishiguro N, Ohno K. Zonisamide enhances neurite elongation of primary motor neurons and facilitates peripheral nerve regeneration in vitro and in a mouse model. PLoS One 2016, 11: e0148470.
    2. Imai K, Kotani T, Tsuda H, Mano Y, Nakano T, Ushida T, Li H, Miki R, Sumigama S, Iwase A, Hirakawa A, Ohno K, Toyokuni S, Takeuchi H, Mizuno T, Suzumura A, Kikkawa F. Neuroprotective potential of molecular hydrogen against perinatal brain injury via suppression of activated microglia. Free Radic Biol Med 2016, 91: 154-163.
    3. Hasegawa S, Kitoh H, Ohkawara B, Mishima K, Matsushita M, Masuda A, Ishiguro N, Ohno K. Tranilast stimulates endochondral ossification by upregulating SOX9 and RUNX2 promoters. Biochem Biophys Res Commun, 2016, 470: 356-361.
    4. Gao K, Wang J, Li L, Zhai Y, Ren Y, You H, Wang B, Wu X, Li J, Liu Z, Li X, Huang Y, Luo XP, Hu D, Ohno K, Wang C. Polymorphisms in four genes (rs151290, rs972283, rs780094 and rs10830963) and their correlation with type 2 diabetes mellitus in Han Chinese in Henan Province, China. Int J Env Res Public Health 2016, 13.
    5. Takegami Y, Ohkawara B, Ito M, Masuda A, Nakashima H, Ishiguro N, Ohno K. R-spondin 2 facilitates differentiation of proliferating chondrocytes into hypertrophic chondrocytes by enhancing Wnt/beta-catenin signaling in endochondral ossification. Biochem Biophys Res Commun 2016, 473: 255-264.
    6. Chen G, Masuda A, Konishi H, Ohkawara B, Ito M, Kinoshita M, Kiyama H, Matsuura T, Ohno K. Phenylbutazone induces expression of MBNL1 and suppresses formation of MBNL1-CUG RNA foci in a mouse model of myotonic dystrophy. Sci Rep 2016, 6: 25317.
    7. Hirayama M, Tsunoda M, Yamamoto M, Tsuda T, Ohno K. Serum tyrosine-to-phenylalanine ratio is low in Parkinson's disease. J Parkinsons Dis 2016, 6: 423-431.
    8. Nakashima H*, Ohkawara B*, Ishigaki S, Fukudome T, Ito K, Tsushima M, Konishi H, Okuno T, Yoshimura T, Ito M, Masuda A, Sobue G, Kiyama H, Ishiguro N, Ohno K. R-spondin 2 promotes acetylcholine receptor clustering at the neuromuscular junction via Lgr5. Sci Rep 2016, 6: 28512. *Equal contribution.
    9. Bruun GH, Doktor TK, Borch J-J, Masuda A, Krainer AR, Ohno K, Andresen BS. Global identification of hnRNP A1 binding sites for SSO-based splicing modulation. BMC Biol 2016, 14: 54.
    10. Shibata A, Okuno T, Rahman MA, Azuma Y, Takeda J, Masuda A, Selcen D, Engel AG, Ohno K. IntSplice: prediction of the splicing consequences of intronic single-nucleotide variations in the human genome. J Hum Genet 2016, 61: 633-640.
    11. Muramatsu Y, Ito M, Oshima T, Kojima S, Ohno K. Hydrogen-rich water ameliorates bronchopulmonary dysplasia (BPD) in newborn rats. Pediatr Pulmonol 2016, 51: 928-935.
    12. Lin Y, Ohkawara B, Ito M, Misawa N, Miyamoto K, Takegami Y, Masuda A, Toyokuni S, Ohno K. Molecular hydrogen suppresses activated Wnt/beta-catenin signaling. Sci Rep 2016, 6: 31986.
    13. Shen X-M*, Okuno T*, Milone M, Otsuka K, Takahashi K, Komaki H, Giles E, Ohno K, Engel AG. Mutations causing slow-channel myasthenia reveal that a valine ring in the channel pore of muscle AChR is optimized for stabilizing channel gating. Hum Mutat, 2016, 37: 1051-1059. *Equal contribution.
    14. Ushida T, Kotani T, Tsuda H, Imai K, Nakano T, Hirako S, Ito Y, Li H, Mano Y, Wang J, Miki R, Yamamoto E, Iwase A, Bando YK, Hirayama M, Ohno K, Toyokuni S, Kikkawa F. Molecular hydrogen ameliorates several characteristics of preeclampsia in the Reduced Uterine Perfusion Pressure (RUPP) rat model. Free Radic Biol Med 2016, 101: 524-533.
    15. Masuda A, Ohno K. Neurodegeneration-associated RNA-binding protein, FUS, regulates mRNA length. Atlas of Science. Ed. by Lynn C Yeoman. AoS Nordic AB, Stockholm, 2016, http://atlasofscience.org/neurodegeneration-associated-rna-binding-protein-fus-regulates-mrna-length/ (査読有)
    16. Ohno K, Yagi H, Ohkawara B. Repositioning again of zonisamide for nerve regeneration. Neural Regener Res 2016, 11: 541-542. (査読有)
    17. Ohno K. Is the serum creatine kinase level elevated in congenital myasthenic syndrome? J Neurol Neurosurg Psychiatry 2016, 87: 801. (査読有)
    18. Ohno K, Ohkawara B, Ito M. Recent advances in congenital myasthenic syndromes. Clin Exp Neuroimmunol 2016, 7: 246–259. (査読有)
    19. Masuda A, Takeda J, Ohno K. FUS-mediated regulation of alternative RNA processing in neurons: insights from global transcriptome analysis. Wiley Interdiscip Rev RNA 2016, 7: 330-340. (査読有)
    20. Ohno K, Otsuka K, Ito M. Roles of collagen Q in MuSK antibody-positive myasthenia gravis. Chem Biol Interact 2016, 259: 266-270. (査読有)
  • 2015
    1. Azuma Y, Nakata T, Tanaka M, Shen XM, Ito M, Iwata S, Okuno T, Nomura Y, Ando N, Ishigaki K, Ohkawara B, Masuda A, Natsume J, Kojima S, Sokabe M, Ohno K. Congenital myasthenic syndrome in Japan: ethnically unique mutations in muscle nicotinic acetylcholine receptor subunits. Neuromuscul Disord, 2015; 25: 60-69.
    2. Matsushita M, Hasegawa S, Kitoh H, Mori K, Ohkawara B, Yasoda A, Masuda A, Ishiguro N, Ohno K. Meclozine promotes longitudinal skeletal growth in transgenic mice with achondroplasia carrying a gain-of-function mutation in the FGFR3 gene. Endocrinology, 2015; 156: 548-554.
    3. Tsunoda M, Hirayama M, Tsuda T, Ohno K. Noninvasive monitoring of plasma L-dopa concentrations using sweat samples in Parkinson's disease. Clin Chim Acta, 2015; 442: 52-55.
    4. Sobue S, Yamai K, Ito M, Ohno K, Ito M, Iwamoto T, Qiao S, Ohkuwa T, Ichihara M. Simultaneous oral and inhalational intake of molecular hydrogen additively suppresses signaling pathways in rodents. Mol Cell Biochem, 2015; 403: 231-241.
    5. Funayama M, Ohe K, Amo T, Furuya N, Yamaguchi J, Saiki S, Li Y, Ogaki K, Ando M, Yoshino H, Tomiyama H, Nishioka K, Hasegawa K, Saiki H, Satake W, Mogushi K, Sasaki R, Kokubo Y, Kuzuhara S, Toda T, Mizuno Y, Uchiyama Y, Ohno K, Hattori N. CHCHD2 mutations in autosomal dominant late-onset Parkinson's disease: a genome-wide linkage and sequencing study. Lancet Neurol, 2015; 14: 274-282.
    6. Selcen D, Ohkawara B, Shen XM, McEvoy K, Ohno K, Engel AG. Impaired Synaptic Development, Maintenance, and Neuromuscular Transmission in LRP4-Related Myasthenia. JAMA Neurol, 2015; 72: 889-896.
    7. Udagawa T, Fujioka Y, Tanaka M, Honda D, Yokoi S, Riku Y, Ibi D, Nagai T, Yamada K, Watanabe H, Katsuno M, Inada T, Ohno K, Sokabe M, Okado H, Ishigaki S, Sobue G. FUS regulates AMPA receptor function and FTLD/ALS-associated behaviour via GluA1 mRNA stabilization. Nat Commun, 2015; 6: 7098.
    8. Fujii H, Matsubara K, Sakai K, Ito M, Ohno K, Ueda M, Yamamoto A. Dopaminergic differentiation of stem cells from human deciduous teeth and their therapeutic benefits for Parkinsonian rats. Brain Res, 2015; 1613: 59-72.
    9. Iwata S, Ito M, Nakata T, Noguchi Y, Okuno T, Ohkawara B, Masuda A, Goto T, Adachi M, Osaka H, Nonaka R, Arikawa-Hirasawa E, Ohno K. A missense mutation in domain III in HSPG2 in Schwartz-Jampel syndrome compromises secretion of perlecan into the extracellular space. Neuromuscul Disord, 2015; 25: 667-671.
    10. Kishimoto Y, Kato T, Ito M, Azuma Y, Fukasawa Y, Ohno K, Kojima S. Hydrogen ameliorates pulmonary hypertension in rats by anti-inflammatory and antioxidant effects. J Thorac Cardiovasc Surg, 2015; 150: 645-654 e643.
    11. Rahman MA, Azuma Y, Nasrin F, Takeda J, Nazim M, Bin Ahsan K, Masuda A, Engel AG, Ohno K. SRSF1 and hnRNP H antagonistically regulate splicing of COLQ exon 16 in a congenital myasthenic syndrome. Sci Rep, 2015; 5: 13208.
    12. Masuda A, Takeda J, Okuno T, Okamoto T, Ohkawara B, Ito M, Ishigaki S, Sobue G, Ohno K. Position-specific binding of FUS to nascent RNA regulates mRNA length. Genes Dev, 2015; 29: 1045-1057.
    13. Otsuka K, Ito M, Ohkawara B, Masuda A, Kawakami Y, Sahashi K, Nishida H, Mabuchi N, Takano A, Engel AG, Ohno K. Collagen Q and anti-MuSK autoantibody competitively suppress agrin/LRP4/MuSK signaling. Sci Rep, 2015; 5: 13928.
    14. Hasegawa S, Goto S, Tsuji H, Okuno T, Asahara T, Nomoto K, Shibata A, Fujisawa Y, Minato T, Okamoto A, Ohno K, Hirayama M. Intestinal Dysbiosis and Lowered Serum Lipopolysaccharide-Binding Protein in Parkinson's Disease. PLoS One, 2015; 10: e0142164.
    15. Yagi H, Ohkawara B, Nakashima H, Ito K, Tsushima M, Ishii H, Noto K, Ohta K, Masuda A, Imagama S, Ishiguro N, Ohno K. Zonisamide Enhances Neurite Elongation of Primary Motor Neurons and Facilitates Peripheral Nerve Regeneration In Vitro and in a Mouse Model. PLoS One, 2015; 10: e0142786.
    16. Mishima K, Kitoh H, Ohkawara B, Okuno T, Ito M, Masuda A, Ishiguro N, Ohno K. Lansoprazole upregulates polyubiquitination of TNF receptor associated factor 6 and facilitates Runx2-mediated osteoblastogenesis. EBioMedicine, 2015; 2: 2046-2061.
    17. Ito M, Ohno K. A hereditary mutation in Schwartz-Jampel syndrome. In: Atlas of Science, edited by LC Yeoman, 2015, AoS Nordic AB, Stockholm.
    18. Rahman MA, Ohno K. Splicing aberrations in congenital myasthenic syndromes. J Investig Genomics, 2015; 2: 00038.
    19. Rahman MA, Nasrin F, Masuda A, Ohno K. Decoding abnormal splicing code in human diseases. J Investig Genomics 2015; 2: 00016
    20. Ichihara M, Sobue S, Ito M, Ito M, Hirayama M, Ohno K. Beneficial biological effects and the underlying mechanisms of molecular hydrogen - comprehensive review of 321 original articles. Med Gas Res, 2015; 5: 12.
  • 2014
    1. Ohkawara B, Cabrera-Serrano M, Nakata T, Milone M, Asai N, Ito K, Ito M, Masuda A, Ito Y, Engel AG, Ohno K. LRP4 third beta-propeller domain mutations cause novel congenital myasthenia by compromising agrin-mediated MuSK signaling in a position-specific manner. Hum Mol Genet, 2014; 23: 1856-1868.
    2. Inaguma Y, Hamada N, Tabata H, Iwamoto I, Mizuno M, Nishimura YV, Ito H, Morishita R, Suzuki M, Ohno K, Kumagai T, Nagata K. SIL1, a causative cochaperone gene of Marinesco-Sojgren syndrome, plays an essential role in establishing the architecture of the developing cerebral cortex. EMBO Mol Med, 2014; 6: 414-429.
    3. Nakayama T, Nakamura H, Oya Y, Kimura T, Imahuku I, Ohno K, Nishino I, Abe K, Matsuura T. Clinical and genetic analysis of the first known Asian family with myotonic dystrophy type 2. J Hum Genet, 2014; 59: 129-133.
    4. Kokunai Y, Nakata T, Furuta M, Sakata S, Kimura H, Aiba T, Yoshinaga M, Osaki Y, Nakamori M, Itoh H, Sato T, Kubota T, Kadota K, Shindo K, Mochizuki H, Shimizu W, Horie M, Okamura Y, Ohno K, Takahashi MP. A Kir3.4 mutation causes Andersen-Tawil syndrome by an inhibitory effect on Kir2.1. Neurology, 2014; 82: 1058-1064.
    5. Mano Y, Kotani T, Ito M, Nagai T, Ichinohashi Y, Yamada K, Ohno K, Kikkawa F, Toyokuni S. Maternal molecular hydrogen administration ameliorates rat fetal hippocampal damage caused by in utero ischemia-reperfusion. Free Radic Biol Med, 2014; 69: 324-330.
    6. Takamatsu A, Ohkawara B, Ito M, Masuda A, Sakai T, Ishiguro N, Ohno K. Verapamil protects against cartilage degradation in osteoarthritis by inhibiting Wnt/beta-catenin signaling. PLoS One, 2014; 9: e92699.
    7. Kobayashi M, Ohno T, Ihara K, Murai A, Kumazawa M, Hoshino H, Iwanaga K, Iwai H, Hamana Y, Ito M, Ohno K, Horio F. Searching for genomic region of high-fat diet-induced type 2 diabetes in mouse chromosome 2 by analysis of congenic strains. PLoS One, 2014; 9: e96271.
    8. Yamashita Y, Matsuura T, Kurosaki T, Amakusa Y, Kinoshita M, Ibi T, Sahashi K, Ohno K. LDB3 splicing abnormalities are specific to skeletal muscles of patients with myotonic dystrophy type 1 and alter its PKC binding affinity. Neurobiol Dis, 2014; 69: 200-205.
    9. Asai N, Ohkawara B, Ito M, Masuda A, Ishiguro N, Ohno K. LRP4 induces extracellular matrix productions and facilitates chondrocyte differentiation. Biochem Biophys Res Commun, 2014; 451: 302-307.
    10. Nasrin F, Rahman MA, Masuda A, Ohe K, Takeda J, Ohno K. HnRNP C, YB-1 and hnRNP L coordinately enhance skipping of human MUSK exon 10 to generate a Wnt-insensitive MuSK isoform. Sci Rep, 2014; 4: 6841.
    11. Ohno K, Ohkawara B, Ito M, Engel AG. Molecular Genetics of Congenital Myasthenic Syndromes. In: eLS, 2014, John Wiley & Sons, Inc.
    12. Ohno K, Ito M, Kawakami Y, Ohtsuka K. Collagen Q is a key player for developing rational therapy for congenital myasthenia and for dissecting the mechanisms of anti-MuSK myasthenia gravis. J Mol Neurosci, 2014; 53: 359-361.
    13. Ohno K. Mutation analysis of a large cohort of GNE myopathy reveals a diverse array of GNE mutations affecting sialic acid biosynthesis. J Neurol Neurosurg Psychiatry, 2014; 85: 831.
    14. Noda M, Ito M, Ohsawa I, Ohno K. Beneficial effects of hydrogen in the CNS and a new brain-stomach interaction. Eur J Neurodeg Dis, 2014; 3: 25-34.
    15. Noda M, Fujita K, Ohsawa I, Ito M, Ohno K. Multiple effects of molecular hydrogen and its distinct mechanism. J Neurol Disord, 2014; 2: 6.
  • 2013
    1. Yamamoto R, Matsushita M, Kitoh H, Masuda A, Ito M, Katagiri T, Kawai T, Ishiguro N, Ohno K. Clinically applicable antianginal agents suppress osteoblastic transformation of myogenic cells and heterotopic ossifications in mice. J Bone Miner Metab, 2013; 31: 26-33.
    2. Sayeed S, Asano E, Ito S, Ohno K, Hamaguchi M, Senga T. S100A10 is required for the organization of actin stress fibers and promotion of cell spreading. Mol Cell Biochem, 2013; 374: 105-111.
    3. Iio A, Ito M, Itoh T, Terazawa R, Fujita Y, Nozawa Y, Ohsawa I, Ohno K, Ito M. Molecular hydrogen attenuates fatty acid uptake and lipid accumulation through downregulating CD36 expression in HepG2 cells. Med Gas Res, 2013; 3: 6.
    4. Nakata T, Ito M, Azuma Y, Otsuka K, Noguchi Y, Komaki H, Okumura A, Shiraishi K, Masuda A, Natsume J, Kojima S, Ohno K. Mutations in the C-terminal domain of ColQ in endplate acetylcholinesterase deficiency compromise ColQ-MuSK interaction. Hum Mutat, 2013; 34: 997-1004.
    5. Tanisawa K, Mikami E, Fuku N, Honda Y, Honda S, Ohsawa I, Ito M, Endo S, Ihara K, Ohno K, Kishimoto Y, Ishigami A, Maruyama N, Sawabe M, Iseki H, Okazaki Y, Hasegawa-Ishii S, Takei S, Shimada A, Hosokawa M, Mori M, Higuchi K, Takeda T, Higuchi M, Tanaka M. Exome sequencing of senescence-accelerated mice (SAM) reveals deleterious mutations in degenerative disease-causing genes. BMC Genomics, 2013; 14: 248.
    6. Selcen D, Shen XM, Milone M, Brengman J, Ohno K, Deymeer F, Finkel R, Rowin J, Engel AG. GFPT1-myasthenia: clinical, structural, and electrophysiologic heterogeneity. Neurology, 2013; 81: 370-378.
    7. Tsunoda M, Hirayama M, Ohno K, Tsuda T. A simple analytical method involving the use of a monolithic silica disk-packed spin column and HPLC-ECD for determination of L-DOPA in plasma of patients with Parkinson's disease. Analytical Methods, 2013; 5: 5161-5164.
    8. Fujioka Y, Ishigaki S, Masuda A, Iguchi Y, Udagawa T, Watanabe H, Katsuno M, Ohno K, Sobue G. FUS-regulated region- and cell-type-specific transcriptome is associated with cell selectivity in ALS/FTLD. Sci Rep, 2013; 3: 2388.
    9. Kitoh H, Achiwa M, Kaneko H, Mishima K, Matsushita M, Kadono I, Horowitz JD, Sallustio BC, Ohno K, Ishiguro N. Perhexiline maleate in the treatment of fibrodysplasia ossificans progressiva: an open-labeled clinical trial. Orphanet J Rare Dis, 2013; 8: 163.
    10. Matsushita M, Kitoh H, Ohkawara B, Mishima K, Kaneko H, Ito M, Masuda A, Ishiguro N, Ohno K. Meclozine facilitates proliferation and differentiation of chondrocytes by attenuating abnormally activated FGFR3 signaling in achondroplasia. PLoS One, 2013; 8: e81569.
    11. Rahman MA, Masuda A, Ohe K, Ito M, Hutchinson DO, Mayeda A, Engel AG, Ohno K. HnRNP L and hnRNP LL antagonistically modulate PTB-mediated splicing suppression of CHRNA1 pre-mRNA. Sci Rep, 2013; 3: 2931.
    12. Honda D, Ishigaki S, Iguchi Y, Fujioka Y, Udagawa T, Masuda A, Ohno K, Katsuno M, Sobue G. The ALS/FTLD-related RNA-binding proteins TDP-43 and FUS have common downstream RNA targets in cortical neurons. FEBS Open Bio, 2013; 4: 1-10.
    13. Ohe K, Masuda A, Ohno K. Intronic and exonic nucleotide variations that affect RNA splicing in humans. In: Genomics I – Humans, Animals and Plants. pp. 29-46, 2013, iConcept Press.
    14. Ohno K, Ito M, Kawakami Y, Krejci E, Engel AG. Specific binding of collagen Q to the neuromuscular junction is exploited to cure congenital myasthenia and to explore bases of myasthenia gravis. Chem Biol Interact, 2013; 203: 335-340.
    15. Ohno K. Glycosylation defects as an emerging novel cause leading to a limb-girdle type of congenital myasthenic syndromes. J Neurol Neurosurg Psychiatry, 2013; 84: 1064.
  • 2012
    1. Masuda A, Andersen HS, Doktor TK, Okamoto T, Ito M, Andresen BS, Ohno K. CUGBP1 and MBNL1 preferentially bind to 3' UTRs and facilitate mRNA decay. Sci Rep, 2012; 2: 209.
    2. Yoshinaga H, Sakoda S, Good JM, Takahashi MP, Kubota T, Arikawa-Hirasawa E, Nakata T, Ohno K, Kitamura T, Kobayashi K, Ohtsuka Y. A novel mutation in SCN4A causes severe myotonia and school-age-onset paralytic episodes. J Neurol Sci, 2012; 315: 15-19.
    3. Ito M, Suzuki Y, Okada T, Fukudome T, Yoshimura T, Masuda A, Takeda S, Krejci E, Ohno K. Protein-anchoring strategy for delivering acetylcholinesterase to the neuromuscular junction. Mol Ther, 2012; 20: 1384-1392.
    4. Ito M, Hirayama M, Yamai K, Goto S, Ito M, Ichihara M, Ohno K. Drinking hydrogen water and intermittent hydrogen gas exposure, but not lactulose or continuous hydrogen gas exposure, prevent 6-hydorxydopamine-induced Parkinson's disease in rats. Med Gas Res, 2012; 2: 15.
    5. Kurosaki T, Ueda S, Ishida T, Abe K, Ohno K, Matsuura T. The unstable CCTG repeat responsible for myotonic dystrophy type 2 originates from an AluSx element insertion into an early primate genome. PLoS One, 2012; 7: e38379.
    6. Ishigaki S, Masuda A, Fujioka Y, Iguchi Y, Katsuno M, Shibata A, Urano F, Sobue G, Ohno K. Position-dependent FUS-RNA interactions regulate alternative splicing events and transcriptions. Sci Rep, 2012; 2: 529.
    7. Yamashita Y, Matsuura T, Shinmi J, Amakusa Y, Masuda A, Ito M, Kinoshita M, Furuya H, Abe K, Ibi T, Sahashi K, Ohno K. Four parameters increase the sensitivity and specificity of the exon array analysis and disclose 25 novel aberrantly spliced exons in myotonic dystrophy. J Hum Genet, 2012; 57: 368-374.
    8. Matsuura T, Minami N, Arahata H, Ohno K, Abe K, Hayashi YK, Nishino I. Myotonic dystrophy type 2 is rare in the Japanese population. J Hum Genet, 2012; 57: 219-220.
    9. Engel AG, Shen X-M, Ohno K, Sine SM. Congenital myasthenic syndromes. In: Myasthenia gravis and myasthenic disorders 2nd ed., edited by AG Engel. pp. 173-230, 2012, Oxford University Press, New York.
    10. Ohno K, Ito M, Engel AG. Congenital Myasthenic Syndromes -Molecular Bases of Congenital Defects of Proteins at the Neuromuscular Junction- In: Neuromuscul Disord. pp. 175-200, 2012, InTech, Rijeka.
    11. Ohno K, Ito M, Ichihara M, Ito M. Molecular hydrogen as an emerging therapeutic medical gas for neurodegenerative and other diseases. Oxid Med Cell Longev, 2012; 2012: 353152.
  • 2011
    1. Selcen D, Juel VC, Hobson-Webb LD, Smith EC, Stickler DE, Bite AV, Ohno K, Engel AG. Myasthenic syndrome caused by plectinopathy. Neurology, 2011; 76: 327-336.
    2. Hirayama M, Nakamura T, Watanabe H, Uchida K, Hama T, Hara T, Niimi Y, Ito M, Ohno K, Sobue G. Urinary 8-hydroxydeoxyguanosine correlate with hallucinations rather than motor symptoms in Parkinson's disease. Parkinsonism Relat Disord, 2011; 17: 46-49.
    3. Fu Y, Masuda A, Ito M, Shinmi J, Ohno K. AG-dependent 3'-splice sites are predisposed to aberrant splicing due to a mutation at the first nucleotide of an exon. Nucleic Acids Res, 2011; 39: 4396-4404.
    4. Itoh T, Hamada N, Terazawa R, Ito M, Ohno K, Ichihara M, Nozawa Y, Ito M. Molecular hydrogen inhibits lipopolysaccharide/interferon gamma-induced nitric oxide production through modulation of signal transduction in macrophages. Biochem Biophys Res Commun, 2011; 411: 143-149.
    5. Kaneko H, Kitoh H, Matsuura T, Masuda A, Ito M, Mottes M, Rauch F, Ishiguro N, Ohno K. Hyperuricemia cosegregating with osteogenesis imperfecta is associated with a mutation in GPATCH8. Hum Genet, 2011; 130: 671-683.
    6. Ito M, Ibi T, Sahashi K, Ichihara M, Ito M, Ohno K. Open-label trial and randomized, double-blind, placebo-controlled, crossover trial of hydrogen-enriched water for mitochondrial and inflammatory myopathies. Med Gas Res, 2011; 1: 24.
    7. Kawakami Y, Ito M, Hirayama M, Sahashi K, Ohkawara B, Masuda A, Nishida H, Mabuchi N, Engel AG, Ohno K. Anti-MuSK autoantibodies block binding of collagen Q to MuSK. Neurology, 2011; 77: 1819-1826.
    8. Ohno K, Engel AG. Molecular defects of acetylcholine receptor subunits in congenital myasthenic syndromes. In: Pharmacology of Nicotinic Acetylcholine Receptors from the Basic and Therapeutic Perspectives, edited by HR Arias. pp. 175-186, 2011, Research Signpost, Kerala.
    9. Ohno K, Masuda A. RNA pathologies in neurological disorders. In: Neurochemical Mechanisms in Disease, edited by JP Blass. pp. 399-415, 2011, Springer, New York.
    10. Ohta S, Nakao A, Ohno K. The 2011 Medical Molecular Hydrogen Symposium: An inaugural symposium of the journal Medical Gas Research. Med Gas Res, 2011; 1: 10.
  • 2009
    1. Fu Y, Ito M, Fujita Y, Ito M, Ichihara M, Masuda A, Suzuki Y, Maesawa S, Kajita Y, Hirayama M, Ohsawa I, Ohta S, Ohno K. Molecular hydrogen is protective against 6-hydroxydopamine-induced nigrostriatal degeneration in a rat model of Parkinson's disease. Neurosci Lett, 2009; 453: 81-85.
    2. Milone M, Shen XM, Selcen D, Ohno K, Brengman J, Iannaccone ST, Harper CM, Engel AG. Myasthenic syndrome due to defects in rapsyn: Clinical and molecular findings in 39 patients. Neurology, 2009; 73: 228-235.
    3. Bian Y, Masuda A, Matsuura T, Ito M, Okushin K, Engel AG, Ohno K. Tannic acid facilitates expression of the polypyrimidine tract binding protein and alleviates deleterious inclusion of CHRNA1 exon P3A due to an hnRNP H-disrupting mutation in congenital myasthenic syndrome. Hum Mol Genet, 2009; 18: 1229-1237.
    4. Almeida T, Alonso I, Martins S, Ramos EM, Azevedo L, Ohno K, Amorim A, Saraiva-Pereira ML, Jardim LB, Matsuura T, Sequeiros J, Silveira I. Ancestral origin of the ATTCT repeat expansion in spinocerebellar ataxia type 10 (SCA10). PLoS One, 2009; 4: e4553.
    5. Kurosaki T, Matsuura T, Ohno K, Ueda S. Alu-mediated acquisition of unstable ATTCT pentanucleotide repeats in the human ATXN10 gene. Mol Biol Evol, 2009; 26: 2573-2579.
    6. Itoh T, Fujita Y, Ito M, Masuda A, Ohno K, Ichihara M, Kojima T, Nozawa Y, Ito M. Molecular hydrogen suppresses FcepsilonRI-mediated signal transduction and prevents degranulation of mast cells. Biochem Biophys Res Commun, 2009; 389: 651-656.
  • 2008
    1. Saito T, Amakusa Y, Kimura T, Yahara O, Aizawa H, Ikeda Y, Day JW, Ranum LP, Ohno K, Matsuura T. Myotonic dystrophy type 2 in Japan: ancestral origin distinct from Caucasian families. Neurogenetics, 2008; 9: 61-63.
    2. Gao K, Masuda A, Matsuura T, Ohno K. Human branch point consensus sequence is yUnAy. Nucleic Acids Res, 2008; 36: 2257-2267.
    3. Kurosaki T, Matsuura T, Ohno K, Ueda S. Long-range PCR for the diagnosis of spinocerebellar ataxia type 10. Neurogenetics, 2008; 9: 151-152.
    4. Shen XM, Fukuda T, Ohno K, Sine SM, Engel AG. Congenital myasthenia-related AChR delta subunit mutation interferes with intersubunit communication essential for channel gating. J Clin Invest, 2008; 118: 1867-1876.
    5. Ito M, Masuda A, Jinno S, Katagiri T, Krejci E, Ohno K. Viral vector-mediated [corrected] expression of human collagen Q in cultured cells. Chem Biol Interact, 2008; 175: 346-348.
    6. Masuda A, Shen XM, Ito M, Matsuura T, Engel AG, Ohno K. hnRNP H enhances skipping of a nonfunctional exon P3A in CHRNA1 and a mutation disrupting its binding causes congenital myasthenic syndrome. Hum Mol Genet, 2008; 17: 4022-4035.
  • 2007
    1. Masuda A, Hashimoto K, Yokoi T, Doi T, Kodama T, Kume H, Ohno K, Matsuguchi T. Essential role of GATA transcriptional factors in the activation of mast cells. J Immunol, 2007; 178: 360-368.
    2. Ichihara M, Murakumo Y, Masuda A, Matsuura T, Asai N, Jijiwa M, Ishida M, Shinmi J, Yatsuya H, Qiao S, Takahashi M, Ohno K. Thermodynamic instability of siRNA duplex is a prerequisite for dependable prediction of siRNA activities. Nucleic Acids Res, 2007; 35: e123.
    3. Sahashi K, Masuda A, Matsuura T, Shinmi J, Zhang Z, Takeshima Y, Matsuo M, Sobue G, Ohno K. In vitro and in silico analysis reveals an efficient algorithm to predict the splicing consequences of mutations at the 5' splice sites. Nucleic Acids Res, 2007; 35: 5995-6003.
  • 2005
    1. Shen XM, Ohno K, Sine SM, Engel AG. Subunit-specific contribution to agonist binding and channel gating revealed by inherited mutation in muscle acetylcholine receptor M3-M4 linker. Brain, 2005; 128: 345-355.
    2. Ohno K, Tsujino A, Shen XM, Milone M, Engel AG. Spectrum of splicing errors caused by CHRNE mutations affecting introns and intron/exon boundaries. J Med Genet, 2005; 42: e53.
    3. Ohno K, Engel AG. Splicing abnormalities in congenital myasthenic syndromes. Acta Myol, 2005; 24: 50-54.
  • 2004
    1. Selcen D, Ohno K, Engel AG. Myofibrillar myopathy: clinical, morphological and genetic studies in 63 patients. Brain, 2004; 127: 439-451.
    2. Sahashi K, Ibi T, Ohno K, Sahashi K, Nakao N, Kondo H. Progressive myopathy with circulating autoantibody against giantin in the Golgi apparatus. Neurology, 2004; 62: 1891-1893.
    3. Ohno K, Engel AG. Lack of founder haplotype for the rapsyn N88K mutation: N88K is an ancient founder mutation or arises from multiple founders. J Med Genet, 2004; 41: e8.
    4. Kimbell LM, Ohno K, Engel AG, Rotundo RL. C-terminal and heparin-binding domains of collagenic tail subunit are both essential for anchoring acetylcholinesterase at the synapse. J Biol Chem, 2004; 279: 10997-11005.
    5. Cai Y, Cronin CN, Engel AG, Ohno K, Hersh LB, Rodgers DW. Choline acetyltransferase structure reveals distribution of mutations that cause motor disorders. EMBO J, 2004; 23: 2047-2058.
    6. Banwell BL, Ohno K, Sieb JP, Engel AG. Novel truncating RAPSN mutations causing congenital myasthenic syndrome responsive to 3,4-diaminopyridine. Neuromuscul Disord, 2004; 14: 202-207.
    7. Engel AG, Ohno K, Sine SM. Congenital myasthenic syndromes (Chapter 66). In: Myology, edited by AG Engel, C Franzini-Armstrong. pp. 1801-1844, 2004, McGraw Hill, New York.
    8. Sine SM, Engel AG, Wang H-L, Ohno K. Molecular Insights into Acetylcholine Receptor Structure and Function Revealed by Mutations Causing Congenital Myasthenic Syndromes. In: Molecular and Cellular Insights into Ion Channel Biology, edited by RA Maue. pp. 95-119, 2004, Elsevier Science, Amsterdam.
  • 2003
    1. Ohno K, Milone M, Shen XM, Engel AG. A frameshifting mutation in CHRNE unmasks skipping of the preceding exon. Hum Mol Genet, 2003; 12: 3055-3066.
    2. Ohno K, Sadeh M, Blatt I, Brengman JM, Engel AG. E-box mutations in the RAPSN promoter region in eight cases with congenital myasthenic syndrome. Hum Mol Genet, 2003; 12: 739-748.
    3. Tsujino A, Maertens C, Ohno K, Shen XM, Fukuda T, Harper CM, Cannon SC, Engel AG. Myasthenic syndrome caused by mutation of the SCN4A sodium channel. Proc Natl Acad Sci U S A, 2003; 100: 7377-7382.
    4. Shen XM, Ohno K, Tsujino A, Brengman JM, Gingold M, Sine SM, Engel AG. Mutation causing severe myasthenia reveals functional asymmetry of AChR signature cystine loops in agonist binding and gating. J Clin Invest, 2003; 111: 497-505.
    5. Engel AG, Ohno K, Harper CM. Congenital myasthenic syndromes. In: Neuromuscular Disorders of Infancy, Childhood, and Adolescence: A Clinician's Approach, edited by HR Jones, C De Vivo D, BT Darras. pp. 555-574, 2003, Butterworth and Heinemann, Boston.
    6. Sine SM, Wang HL, Ohno K, Shen XM, Lee WY, Engel AG. Mechanistic diversity underlying fast channel congenital myasthenic syndromes. Ann N Y Acad Sci, 2003; 998: 128-137.
    7. Engel AG, Ohno K, Sine SM. Congenital myasthenic syndromes: A diverse array of molecular targets. J Neurocytol, 2003; 32: 1017-1037.
    8. Engel AG, Ohno K, Sine SM. Sleuthing molecular targets for neurological diseases at the neuromuscular junction. Nat Rev Neurosci, 2003; 4: 339-352.
    9. Engel AG, Ohno K, Sine SM. Congenital myasthenic syndromes: progress over the past decade. Muscle Nerve, 2003; 27: 4-25.
    10. Engel AG, Ohno K, Shen XM, Sine SM. Congenital myasthenic syndromes: Multiple molecular targets at the neuromuscular junction. Myasthenia Gravis and Related Disorders, 2003; 998: 138-160.
  • 2002
    1. Shen XM, Ohno K, Fukudome T, Tsujino A, Brengman JM, De Vivo DC, Packer RJ, Engel AG. Congenital myasthenic syndrome caused by low-expressor fast-channel AChR delta subunit mutation. Neurology, 2002; 59: 1881-1888.
    2. Shapira YA, Sadeh ME, Bergtraum MP, Tsujino A, Ohno K, Shen XM, Brengman J, Edwardson S, Matoth I, Engel AG. Three novel COLQ mutations and variation of phenotypic expressivity due to G240X. Neurology, 2002; 58: 603-609.
    3. Ohno K, Engel AG, Shen XM, Selcen D, Brengman J, Harper CM, Tsujino A, Milone M. Rapsyn mutations in humans cause endplate acetylcholine-receptor deficiency and myasthenic syndrome. Am J Hum Genet, 2002; 70: 875-885.
    4. Sine SM, Shen XM, Wang HL, Ohno K, Lee WY, Tsujino A, Brengmann J, Bren N, Vajsar J, Engel AG. Naturally occurring mutations at the acetylcholine receptor binding site independently alter ACh binding and channel gating. J Gen Physiol, 2002; 120: 483-496.
    5. Byring RF, Pihko H, Tsujino A, Shen XM, Gustafsson B, Hackman P, Ohno K, Engel AG, Udd B. Congenital myasthenic syndrome associated with episodic apnea and sudden infant death. Neuromuscul Disord, 2002; 12: 548-553.
    6. Engel AG, Ohno K. Congenital myasthenic syndromes (Chapter 13). In: Adv Neurol, edited by R Pourmand, Y Harati. pp. 203-215, 2002, Lippincott Williams & Wilkins, Philadelphia.
    7. Engel AG, Ohno K, Selcen D. Congenital Myasthenic Syndromes. In: Structural and Molecular Basis of Skeletal Muscle Diseases, edited by G Karpati. pp. 170-179, 2002, International Society of Neuropathology/World Federation of Neurology. ISN Neuropath Press, Basel.
    8. Ohno K, Engel AG. Congenital myasthenic syndromes: genetic defects of the neuromuscular junction. Curr Neurol Neurosci Rep, 2002; 2: 78-88.
    9. Engel AG, Ohno K, Sine SM. The spectrum of congenital myasthenic syndromes. Mol Neurobiol, 2002; 26: 347-367.
  • 2001
    1. Sahashi K, Yoneda M, Ohno K, Tanaka M, Ibi T, Sahashi K. Functional characterisation of mitochondrial tRNA(Tyr) mutation (5877-->GA) associated with familial chronic progressive external ophthalmoplegia. J Med Genet, 2001; 38: 703-705.
    2. Ohno K, Tsujino A, Brengman JM, Harper CM, Bajzer Z, Udd B, Beyring R, Robb S, Kirkham FJ, Engel AG. Choline acetyltransferase mutations cause myasthenic syndrome associated with episodic apnea in humans. Proc Natl Acad Sci U S A, 2001; 98: 2017-2022.
    3. Engel AG, Ohno K, Sine SM. Acetylcholine receptor channelopathies and other congenital myasthenic syndromes (Chapter 12). In: Channelopathies of the nervous system, edited by MR Rose, RC Griggs. pp. 179-191, 2001, Butterworth and Heinemann, Boston.
  • 2000
    1. Ohno K, Engel AG, Brengman JM, Shen XM, Heidenreich F, Vincent A, Milone M, Tan E, Demirci M, Walsh P, Nakano S, Akiguchi I. The spectrum of mutations causing end-plate acetylcholinesterase deficiency. Ann Neurol, 2000; 47: 162-170.
    2. Wang HL, Ohno K, Milone M, Brengman JM, Evoli A, Batocchi AP, Middleton LT, Christodoulou K, Engel AG, Sine SM. Fundamental gating mechanism of nicotinic receptor channel revealed by mutation causing a congenital myasthenic syndrome. J Gen Physiol, 2000; 116: 449-462.
    3. Engel AG, Ohno K, Stans AA. Congenital myasthenic syndromes. In: Neuromuscular Diseases: From Basic Mechanisms To Clinical Management, edited by F Demeer. pp. 96-112, 2000, Karger, Basel.
    4. Engel AG, Ohno K, Shen XM, Milone M, Tsujino A. Congenital myasthenic syndromes in the molecular era. Acta Myol, 2000; 19: 5-21.
  • 1999
    1. Middleton L, Ohno K, Christodoulou K, Brengman J, Milone M, Neocleous V, Serdaroglu P, Deymeer F, Ozdemir C, Mubaidin A, Horany K, Al-Shehab A, Mavromatis I, Mylonas I, Tsingis M, Zamba E, Pantzaris M, Kyriallis K, Engel AG. Chromosome 17p-linked myasthenias stem from defects in the acetylcholine receptor epsilon-subunit gene. Neurology, 1999; 53: 1076-1082.
    2. Ohno K, Brengman JM, Felice KJ, Cornblath DR, Engel AG. Congenital end-plate acetylcholinesterase deficiency caused by a nonsense mutation and an A-->G splice-donor-site mutation at position +3 of the collagenlike-tail-subunit gene (COLQ): how does G at position +3 result in aberrant splicing? Am J Hum Genet, 1999; 65: 635-644.
    3. Wang HL, Milone M, Ohno K, Shen XM, Tsujino A, Batocchi AP, Tonali P, Brengman J, Engel AG, Sine SM. Acetylcholine receptor M3 domain: stereochemical and volume contributions to channel gating. Nat Neurosci, 1999; 2: 226-233.
    4. Quiram PA, Ohno K, Milone M, Patterson MC, Pruitt NJ, Brengman JM, Sine SM, Engel AG. Mutation causing congenital myasthenia reveals acetylcholine receptor beta/delta subunit interaction essential for assembly. J Clin Invest, 1999; 104: 1403-1410.
    5. Ohno K, Anlar B, Engel AG. Congenital myasthenic syndrome caused by a mutation in the Ets-binding site of the promoter region of the acetylcholine receptor epsilon subunit gene. Neuromuscul Disord, 1999; 9: 131-135.
    6. Engel AG, Ohno K, Sine SM. Congenital myasthenic syndromes (Chapter 11). In: Myasthenia gravis and myasthenic disorders, edited by AG Engel. pp. 251-297, 1999, Oxford University Press, New York.
    7. Engel AG, Ohno K, Sine SM. Congenital myasthenic syndromes: recent advances. Arch Neurol, 1999; 56: 163-167.
  • 1998
    1. Ohno K, Anlar B, Ozdirim E, Brengman JM, DeBleecker JL, Engel AG. Myasthenic syndromes in Turkish kinships due to mutations in the acetylcholine receptor. Ann Neurol, 1998; 44: 234-241.
    2. Ohno K, Anlar B, Ozdirim E, Brengman JM, Engel AG. Frameshifting and splice-site mutations in the acetylcholine receptor epsilon subunit gene in three Turkish kinships with congenital myasthenic syndromes. Ann N Y Acad Sci, 1998; 841: 189-194.
    3. Milone M, Ohno K, Fukudome T, Shen XM, Brengman J, Griggs RC, Engel AG. Congenital myasthenic syndrome caused by novel loss-of-function mutations in the human AChR epsilon subunit gene. Ann N Y Acad Sci, 1998; 841: 184-188.
    4. Fukudome T, Ohno K, Brengman JM, Engel AG. AChR channel blockade by quinidine sulfate reduces channel open duration in the slow-channel congenital myasthenic syndrome. Ann N Y Acad Sci, 1998; 841: 199-202.
    5. Ohno K, Brengman J, Tsujino A, Engel AG. Human endplate acetylcholinesterase deficiency caused by mutations in the collagen-like tail subunit (ColQ) of the asymmetric enzyme. Proc Natl Acad Sci U S A, 1998; 95: 9654-9659.
    6. Milone M, Wang HL, Ohno K, Prince R, Fukudome T, Shen XM, Brengman JM, Griggs RC, Sine SM, Engel AG. Mode switching kinetics produced by a naturally occurring mutation in the cytoplasmic loop of the human acetylcholine receptor epsilon subunit. Neuron, 1998; 20: 575-588.
    7. Fukudome T, Ohno K, Brengman JM, Engel AG. Quinidine normalizes the open duration of slow-channel mutants of the acetylcholine receptor. Neuroreport, 1998; 9: 1907-1911.
    8. Ohno K, Engel AG. Congenital myasthenic syndromes: gene mutation. Neuromuscular disorders : NMD, 1998; 8: XII-XIII.
    9. Engel AG, Ohno K, Wang HL, Milone M, Sine SM. Molecular basis of congenital myasthenic syndromes: Mutations in the acetylcholine receptor. Neuroscientist, 1998; 4: 185-194.
    10. Engel AG, Ohno K, Sine SM. Congenital myasthenic syndromes: experiments of nature. J Physiol Paris, 1998; 92: 113-117.
    11. Engel AG, Ohno K, Milone M, Sine SM. Congenital myasthenic syndromes. New insights from molecular genetic and patch-clamp studies. Ann N Y Acad Sci, 1998; 841: 140-156.
  • 1997
    1. Wang HL, Auerbach A, Bren N, Ohno K, Engel AG, Sine SM. Mutation in the M1 domain of the acetylcholine receptor alpha subunit decreases the rate of agonist dissociation. J Gen Physiol, 1997; 109: 757-766.
    2. Milone M, Wang HL, Ohno K, Fukudome T, Pruitt JN, Bren N, Sine SM, Engel AG. Slow-channel myasthenic syndrome caused by enhanced activation, desensitization, and agonist binding affinity attributable to mutation in the M2 domain of the acetylcholine receptor alpha subunit. J Neurosci, 1997; 17: 5651-5665.
    3. Ohno K, Quiram PA, Milone M, Wang HL, Harper MC, Pruitt JN, 2nd, Brengman JM, Pao L, Fischbeck KH, Crawford TO, Sine SM, Engel AG. Congenital myasthenic syndromes due to heteroallelic nonsense/missense mutations in the acetylcholine receptor epsilon subunit gene: identification and functional characterization of six new mutations. Hum Mol Genet, 1997; 6: 753-766.
  • 1996
    1. Engel AG, Ohno K, Milone M, Sine SM. Congenital myasthenic syndromes caused by mutations in acetylcholine receptor genes. Neurology, 1997; 48: 28S-35S.
    2. Sawano T, Tanaka M, Ohno K, Yoneda M, Ota Y, Terasaki H, Awaya S, Ozawa T. Mitochondrial DNA mutations associated with the 11778 mutation in Leber's disease. Biochem Mol Biol Int, 1996; 38: 693-700.
    3. Ohno K, Yamamoto M, Engel AG, Harper CM, Roberts LR, Tan GH, Fatourechi V. MELAS- and Kearns-Sayre-type co-mutation [corrected] with myopathy and autoimmune polyendocrinopathy. Ann Neurol, 1996; 39: 761-766.
    4. Engel AG, Ohno K, Bouzat C, Sine SM, Griggs RC. End-plate acetylcholine receptor deficiency due to nonsense mutations in the epsilon subunit. Ann Neurol, 1996; 40: 810-817.
    5. Ohno K, Wang HL, Milone M, Bren N, Brengman JM, Nakano S, Quiram P, Pruitt JN, Sine SM, Engel AG. Congenital myasthenic syndrome caused by decreased agonist binding affinity due to a mutation in the acetylcholine receptor epsilon subunit. Neuron, 1996; 17: 157-170.
    6. Engel AG, Ohno K, Milone M, Wang HL, Nakano S, Bouzat C, Pruitt JN, 2nd, Hutchinson DO, Brengman JM, Bren N, Sieb JP, Sine SM. New mutations in acetylcholine receptor subunit genes reveal heterogeneity in the slow-channel congenital myasthenic syndrome. Hum Mol Genet, 1996; 5: 1217-1227.
  • 1995
    1. Ohno K, Hutchinson DO, Milone M, Brengman JM, Bouzat C, Sine SM, Engel AG. Congenital myasthenic syndrome caused by prolonged acetylcholine receptor channel openings due to a mutation in the M2 domain of the epsilon subunit. Proc Natl Acad Sci U S A, 1995; 92: 758-762.
    2. Sine SM, Ohno K, Bouzat C, Auerbach A, Milone M, Pruitt JN, Engel AG. Mutation of the acetylcholine receptor alpha subunit causes a slow-channel myasthenic syndrome by enhancing agonist binding affinity. Neuron, 1995; 15: 229-239.
  • 1993
    1. Sano T, Ban K, Ichiki T, Kobayashi M, Tanaka M, Ohno K, Ozawa T. Molecular and genetic analyses of two patients with Pearson's marrow-pancreas syndrome. Pediatr Res, 1993; 34: 105-110.
    2. Suoh H, Sahashi K, Ibi T, Tashiro M, Tanaka F, Mitsuma T, Ohno K. Progressive external ophthalmoplegia and myositis. Intern Med, 1993; 32: 319-322.
  • 1992
    1. Sahashi K, Tanaka M, Tashiro M, Ohno K, Ibi T, Takahashi A, Ozawa T. Increased mitochondrial DNA deletions in the skeletal muscle of myotonic dystrophy. Gerontology, 1992; 38: 18-29.
  • 1991
    1. Ohno K, Tanaka M, Sahashi K, Ibi T, Sato W, Yamamoto T, Takahashi A, Ozawa T. Mitochondrial DNA deletions in inherited recurrent myoglobinuria. Ann Neurol, 1991; 29: 364-369.
    2. Tanaka M, Ino H, Ohno K, Ohbayashi T, Ikebe S, Sano T, Ichiki T, Kobayashi M, Wada Y, Ozawa T. Mitochondrial DNA mutations in mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS). Biochem Biophys Res Commun, 1991; 174: 861-868.
    3. Ozawa T, Tanaka M, Ino H, Ohno K, Sano T, Wada Y, Yoneda M, Tanno Y, Miyatake T, Tanaka T, et al. Distinct clustering of point mutations in mitochondrial DNA among patients with mitochondrial encephalomyopathies and with Parkinson's disease. Biochem Biophys Res Commun, 1991; 176: 938-946.
    4. Ozawa T, Tanaka M, Sugiyama S, Ino H, Ohno K, Hattori K, Ohbayashi T, Ito T, Deguchi H, Kawamura K, et al. Patients with idiopathic cardiomyopathy belong to the same mitochondrial DNA gene family of Parkinson's disease and mitochondrial encephalomyopathy. Biochem Biophys Res Commun, 1991; 177: 518-525.
    5. Ohno K, Tanaka M, Suzuki H, Ohbayashi T, Ikebe S, Ino H, Kumar S, Takahashi A, Ozawa T. Identification of a possible control element, Mt5, in the major noncoding region of mitochondrial DNA by intraspecific nucleotide conservation. Biochem Int, 1991; 24: 263-272.
    6. Ota Y, Tanaka M, Sato W, Ohno K, Yamamoto T, Maehara M, Negoro T, Watanabe K, Awaya S, Ozawa T. Detection of platelet mitochondrial DNA deletions in Kearns-Sayre syndrome. Invest Ophthalmol Vis Sci, 1991; 32: 2667-2675.
    7. Ino H, Tanaka M, Ohno K, Hattori K, Ikebe S, Sano T, Ozawa T, Ichiki T, Kobayashi M, Wada Y. Mitochondrial leucine tRNA mutation in a mitochondrial encephalomyopathy. Lancet, 1991; 337: 234-235.
    8. Ohno K, Tanaka M, Ino H, Suzuki H, Tashiro M, Ibi T, Sahashi K, Takahashi A, Ozawa T. Direct DNA sequencing from colony: analysis of multiple deletions of mitochondrial genome. Biochim Biophys Acta, 1991; 1090: 9-16.
    9. Ozawa T, Tanaka M, Hayakawa M, Sugiyama S, Ino H, Sato W, Ohno K, Ikebe S, Yoneda M. Mitochondrial DNA disease: phylogeny and expression. In: New Era of Bioenergetics, edited by Y Mukohata. pp. 247-272, 1991, Academic Press, Tokyo.
    10. Ozawa T, Tanaka M, Hayakawa M, Sugiyama S, Sato W, Ohno K, Ikebe S, Yoneda M. Mitochondrial DNA mutations: types, mechanism and expression. In: Progress in Neuropathology Vol. 7, Mitochondrial Encephalomyopathies, edited by T Sato. pp. 141-151, 1991, Raven Press, New York.
    11. Ozawa T, Tanaka M, Sato W, Ohno K, Yoneda M. Diseases caused by mitochondrial DNA mutations: types and mechanism. In: Proceedings of the XIth International Congress of Neuropathology. pp. 481-485, 1991, Jpn. Soc. Neuropathol., Tokyo.
    12. Ozawa T, Tanaka M, Sato W, Ohno K, Yoneda M, Yamamoto T. Types and mechanism of mitochondrial DNA mutations in mitochondrial myopathy and related diseases. In: Molecular Basis of Neurological Disorders and their Treatment, edited by JW Gorrod, O Albano, E Ferrari, S Papa. pp. 173-190, 1991, Chapman and Hall, London.
    13. Sahashi K, Ibi T, Ohno K, Tanaka M, Tashiro M, Tsuchiya I, Nakao M, Yuasa K, Mitsuma T, Takahashi A, Ozawa T. Visceral myopathy with external ophthalmoplegia and multiple mitochondrial DNA deletions. In: New Trends in Autonomic Nervous System Research, edited by Mea Yoshikawa. pp. 229-230, 1991, Elsevier Science Publishers, B. V.
    14. Tanaka M, Hattori K, Ito H, Ohbayashi T, Ohno K, Sato W, Sugiyama S, Ozawa T. Mitochondrial DNA mutations in idiopathic cardiomyopathy and in presbycardia. In: Mitochondrial Encephalomyopathies, edited by T Sato. pp. 225-236, 1991, Raven Press, New York.
  • 1990
    1. Ozawa T, Tanaka M, Sugiyama S, Hattori K, Ito T, Ohno K, Takahashi A, Sato W, Takada G, Mayumi B, et al. Multiple mitochondrial DNA deletions exist in cardiomyocytes of patients with hypertrophic or dilated cardiomyopathy. Biochem Biophys Res Commun, 1990; 170: 830-836.
    2. Ikebe S, Tanaka M, Ohno K, Sato W, Hattori K, Kondo T, Mizuno Y, Ozawa T. Increase of deleted mitochondrial DNA in the striatum in Parkinson's disease and senescence. Biochem Biophys Res Commun, 1990; 170: 1044-1048.
    3. Ozawa T, Tanaka M, Ikebe S, Ohno K, Kondo T, Mizuno Y. Quantitative determination of deleted mitochondrial DNA relative to normal DNA in parkinsonian striatum by a kinetic PCR analysis. Biochem Biophys Res Commun, 1990; 172: 483-489.
    4. Sahashi K, Ohno K, Tanaka M, Ibi T, Yamamoto T, Tashiro M, Sato W, Takahashi A, Ozawa T. Cytoplasmic body and mitochondrial DNA deletion. J Neurol Sci, 1990; 99: 291-300.
    5. Tanaka M, Ino H, Ohno K, Hattori K, Sato W, Ozawa T, Tanaka T, Itoyama S. Mitochondrial mutation in fatal infantile cardiomyopathy. Lancet, 1990; 336: 1452.
    6. Ozawa T, Tanaka M, Sato W, Ohno K, Sugiyama S, Yoneda M, Yamamoto T, Hattori K, Ikebe S, Tashiro M, Sahashi K. Mitochondrial DNA mutations as an etiology of human degenerative diseases. In: Bioenergetics: Molecular Biology, Biochemistry, and Pathology, edited by CH Kim, T Ozawa. pp. 413-427, 1990, Plenum, New York and London.
    7. Tanaka M, ., Sato W, Ohno K, Yamamoto T, Ozawa T. S1 nuclease analysis and direct sequencing of deleted mitochondrial DNA in myopathic patients: Role of directly repeated sequences in deletion. In: Bioenergetics: Molecular Biology, Biochemistry, and Pathology, edited by CH Kim, T Ozawa. pp. 441-449, 1990, Plenum, New York.
  • 1989
    1. Sato W, Tanaka M, Ohno K, Yamamoto T, Takada G, Ozawa T. Multiple populations of deleted mitochondrial DNA detected by a novel gene amplification method. Biochem Biophys Res Commun, 1989; 162: 664-672.
    2. Tanaka M, Sato W, Ohno K, Yamamoto T, Ozawa T. Direct sequencing of deleted mitochondrial DNA in myopathic patients. Biochem Biophys Res Commun, 1989; 164: 156-163.
    3. Tanaka-Yamamoto T, Tanaka M, Ohno K, Sato W, Horai S, Ozawa T. Specific amplification of deleted mitochondrial DNA from a myopathic patient and analysis of deleted region with S1 nuclease. Biochim Biophys Acta, 1989; 1009: 151-155.
    4. Tanaka M, Yoneda M, Ohno K, Sato W, Yamamoto M, Nonaka I, Horai S, Ozawa T. Differently deleted mitochondrial genomes in maternally inherited chronic progressive external ophthalmoplegia. J Inherit Metab Dis, 1989; 12: 359-362.
  • 1988
    1. Ozawa T, Yoneda M, Tanaka M, Ohno K, Sato W, Suzuki H, Nishikimi M, Yamamoto M, Nonaka I, Horai S. Maternal inheritance of deleted mitochondrial DNA in a family with mitochondrial myopathy. Biochem Biophys Res Commun, 1988; 154: 1240-1247.

Research Keywords

Congenital myasthenic syndromes、 neuromuscular junction、 pre-mRNA splicing、 RNA-binding proteins、 drug repositioning、 Parkinson's disease、 intestinal microbiota、 molecular hydrogen

Call for Graduate Students
We are not taking new students now.