Network


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

Hotspot


Dive into the research topics where Jun Kudoh is active.

Publication


Featured researches published by Jun Kudoh.


Nature Genetics | 2009

A homozygous frameshift mutation in the mouse Flg gene facilitates enhanced percutaneous allergen priming

Padraic G. Fallon; Takashi Sasaki; Aileen Sandilands; Linda E. Campbell; Sean P. Saunders; Niamh E. Mangan; John J. Callanan; Hiroshi Kawasaki; Aiko Shiohama; Akiharu Kubo; John P. Sundberg; Richard B. Presland; Philip Fleckman; Nobuyoshi Shimizu; Jun Kudoh; Alan D. Irvine; Masayuki Amagai; W.H. Irwin McLean

Loss-of-function mutations in the FLG (filaggrin) gene cause the semidominant keratinizing disorder ichthyosis vulgaris and convey major genetic risk for atopic dermatitis (eczema), eczema-associated asthma and other allergic phenotypes. Several low-frequency FLG null alleles occur in Europeans and Asians, with a cumulative frequency of ∼9% in Europe. Here we report a 1-bp deletion mutation, 5303delA, analogous to common human FLG mutations, within the murine Flg gene in the spontaneous mouse mutant flaky tail (ft). We demonstrate that topical application of allergen to mice homozygous for this mutation results in cutaneous inflammatory infiltrates and enhanced cutaneous allergen priming with development of allergen-specific antibody responses. These data validate flaky tail as a useful model of filaggrin deficiency and provide experimental evidence for the hypothesis that antigen transfer through a defective epidermal barrier is a key mechanism underlying elevated IgE sensitization and initiation of cutaneous inflammation in humans with filaggrin-related atopic disease.


Human Genetics | 1998

A common mutation in Sardinian autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy patients

Maria Cristina Rosatelli; Alessandra Meloni; Antonella Meloni; Marcella Devoto; Antonio Cao; Hamish S. Scott; Pärt Peterson; Maarit Heino; Kai Krohn; Kentaro Nagamine; Jun Kudoh; Nobuyoshi Shimizu

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED; also called APS-1,) is a rare autosomal recessive disorder that is more frequent in certain isolated populations. It is characterized by two of the three major clinical symptoms that may be present: Addison’s disease, and/or hypoparathyroidism and/or chronic mucocutaneous candidiasis. We have recently identified the gene for APECED, which we termed AIRE (for autoimmune regulator). AIRE is expressed in thymus, lymph nodes and fetal liver, and encodes a protein with two putative zinc fingers and other motifs suggestive of a transcriptional regulator. Seven mutations have been described to date, including R257X, the predominant Finnish and northern Italian APECED allele, which has also been observed in other patients of diverse origin on different haplotypes. A 13-bp deletion (1094–1106del) has also been observed in several patients of different geo-ethnic origin. The other described mutations appear to be rare. We present mutational analyses of the AIRE gene in ten Sardinian APECED families and show that there is a mutation, R139X, associated with one predominant haplotype unique to the Sardinian patients (18/20 independent alleles). The carrier frequency of R139X in Sardinia is 1.7%, giving an estimated population frequency of APECED of 1/14,400. Using linkage disequilibrium data, the estimated age of the R139X mutation is between 20 and 25 generations. A previously described 13-bp deletion was also observed on an allele of one patient. The identification of a single common Sardinian APECED mutation will facilitate its genetic diagnosis. Given the carrier frequency of R139X in the Sardinian population, AIRE may be implicated in the pathogenesis of other autoimmune diseases in the Sardinian population, particularly those affecting the endocrine system.


European Journal of Immunology | 2000

RNA and protein expression of the murine autoimmune regulator gene (Aire) in normal, RelB-deficient and in NOD mouse.

Maarit Heino; Pärt Peterson; Niko Sillanpää; Sandrine Guérin; Li Wu; Graham Anderson; Hamish S. Scott; Jun Kudoh; Nobuyoshi Shimizu; Eric J. Jenkinson; Philippe Naquet; Kai Krohn

Mutations in the putative transcription factor autoimmune regulator (AIRE) gene are responsible for autoimmune polyendocrinopathy‐candidiosis‐ectodermal dystrophy (APECED; OMIM#240300), a monogenic recessively inherited disease characterized by destructive autoimmune diseases of the endocrine organs, chronic candidiosis of mucous membranes and ectodermal dystrophies. In this study the expression of murine homolog for AIRE protein, Aire, was detected in a fraction of thymic medullary epithelial cells. Subcellularly, in thymus the protein appears as concentrated into nuclear dot‐like structures, whereas in transfected cells the protein is also bound along a cytosolic fibrillar network. By RT‐PCR Aire mRNA was detected in thymus, lymph node, spleen and testis although the second round PCR amplified Aire specific band from most mouse tissues analyzed. Furthermore, the Aire mRNA was detected in dendritic cell (DC) populations isolated from thymus and spleen, representing both myeloid‐ and lymphoid‐related lineages of DC. We also demonstrate that the Aire protein is absent in the thymus of RelB‐deficient mouse and in NOD thymus most of the Aire positive cells showed an abnormal morphology. These results suggest that the Aire protein is associated with the normal development and / or action of a subset of thymic medullary stromal cells involved in tolerance induction.


Nature Genetics | 2001

Insertion of β-satellite repeats identifies a transmembrane protease causing both congenital and childhood onset autosomal recessive deafness

Hamish S. Scott; Jun Kudoh; Marie Wattenhofer; Kazunori Shibuya; Asher Berry; Roman Chrast; Michel Guipponi; Jun Wang; Kazuhiko Kawasaki; Shuichi Asakawa; Shinsei Minoshima; Farah Younus; S. Qasim Mehdi; Uppala Radhakrishna; Marie Pierre Papasavvas; Corinne Gehrig; Colette Rossier; Michael Korostishevsky; Andreas Gal; Nobuyoshi Shimizu; Batsheva Bonne-Tamir

Approximately 50% of childhood deafness is caused by mutations in specific genes. Autosomal recessive loci account for approximately 80% of nonsyndromic genetic deafness. Here we report the identification of a new transmembrane serine protease (TMPRSS3; also known as ECHOS1) expressed in many tissues, including fetal cochlea, which is mutated in the families used to describe both the DFNB10 and DFNB8 loci. An 8-bp deletion and insertion of 18 monomeric (∼68-bp) β-satellite repeat units, normally present in tandem arrays of up to several hundred kilobases on the short arms of acrocentric chromosomes, causes congenital deafness (DFNB10). A mutation in a splice-acceptor site, resulting in a 4-bp insertion in the mRNA and a frameshift, was detected in childhood onset deafness (DFNB8). This is the first description of β-satellite insertion into an active gene resulting in a pathogenic state, and the first description of a protease involved in hearing loss.


Journal of Experimental Medicine | 2004

AIRE Functions As an E3 Ubiquitin Ligase

Daisuke Uchida; Shigetsugu Hatakeyama; Akemi Matsushima; Hongwei Han; Satoshi Ishido; Hak Hotta; Jun Kudoh; Nobuyoshi Shimizu; Vassilis Doucas; Keiichi I. Nakayama; Noriyuki Kuroda; Mitsuru Matsumoto

Autoimmune regulator (AIRE) gene mutation is responsible for the development of autoimmune-polyendocrinopathy-candidiasis ectodermal dystrophy, an organ-specific autoimmune disease with monogenic autosomal recessive inheritance. AIRE is predominantly expressed in medullary epithelial cells of the thymus and is considered to play important roles in the establishment of self-tolerance. AIRE contains two plant homeodomain (PHD) domains, and the novel role of PHD as an E3 ubiquitin (Ub) ligase has just emerged. Here we show that the first PHD (PHD1) of AIRE mediates E3 ligase activity. The significance of this finding was underscored by the fact that disease-causing missense mutations in the PHD1 (C311Y and P326Q) abolished its E3 ligase activity. These results add a novel enzymatic function for AIRE and suggest an indispensable role of the Ub proteasome pathway in the establishment of self-tolerance, in which AIRE is involved.


Immunology Letters | 2002

Expression of AIRE gene in peripheral monocyte/dendritic cell lineage

Kazuhiko Kogawa; Seiho Nagafuchi; Hitoshi Katsuta; Jun Kudoh; Sadafumi Tamiya; Yumiko Sakai; Nobuyoshi Shimizu; Mine Harada

The responsible gene for autoimmune polyglandular syndrome type 1, known as autoimmune regulator (AIRE), was identified by positional cloning. The AIRE gene was reported to be expressed in the thymus medulla and lymph nodes. However, an expression of the AIRE gene in peripheral blood cells has not yet been reported. In the present study, we found that the AIRE gene was restrictively expressed in peripheral CD14-positive monocytes but not in CD4-positive T cells nor polymorphonuclear cells, as assessed by RT-PCR. Moreover, immunocytochemical study revealed the expression of the AIRE protein not only in CD14-positive monocytes but also in differentiated dendritic cells, cultured in RPMI1640 medium containing 800 U/ml GM-CSF, 1000 U/ml IL-4 and 100 U/ml TNF-alpha. Thus, it was concluded that the AIRE gene is restrictively expressed in the peripheral monocyte/dendritic cell lineage.


Human Mutation | 1999

Mutation analyses of North American APS-1 patients

Maarit Heino; Hamish S. Scott; Qiao-Yi Chen; Pärt Peterson; Ulla Mäenpää; Marie Pierre Papasavvas; Christine Barras; Colette Rossier; George P. Chrousos; Constantine A. Stratakis; Kentaro Nagamine; Jun Kudoh; Nobuyoshi Shimizu; Noel Maclaren; Kai Krohn

Autoimmune polyendocrinopathy syndrome type 1 (APS‐1; MIM# 240300) is a rare autosomal recessively inherited disease characterised by destructive autoimmune diseases of endocrine glands. The gene responsible for APS‐1, known as AIRE (for autoimmune regulator), was recently identified and contains motifs suggestive of a transcription regulator. To date, nine APS‐1‐associated mutations have been identified in the AIRE gene, including two common mutations R257X and 1094‐1106del. In addition to these two mutations, we report seven novel mutations in 16 APS‐1 patients from North America. We found that 1094‐1106del and R257X were the most common mutations in this population of mixed geoethnic origin, accounting for 17/32 and 4/32 alleles, respectively. Haplotype analyses suggest that both are recurrent mutations, occurring on several different haplotypes with closely linked markers. All the novel mutations appear to be rare, occurring in only single APS‐1 families. After examining all coding sequences and exon/intron boundaries of the AIRE gene, the other APS‐1 allele remained unidentified in three patients. Genotype‐phenotype correlations for APS‐1 remain difficult, suggesting that other genetic or environmental factors, or both, influence the clinical presentation and disease progression in individual APS‐1 patients. Hum Mutat 13:69–74, 1999.


Current Eye Research | 1998

Spectrum of pathogenic mitochondrial DNA mutations and clinical features in Japanese families with Leber's hereditary optic neuropathy

Yukihiko Mashima; Keiko Yamada; Masato Wakakura; Kazuteru Kigasawa; Jun Kudoh; Nobuyoshi Shimizu; Yoshihisa Oguchi

PURPOSE To investigate the incidence and clinical significance of primary or proposed secondary mitochondrial DNA (mtDNA) mutations in Japanese patients with Lebers hereditary optic neuropathy (LHON). METHODS Blood samples from the 80 unrelated Japanese patients with bilateral optic atrophy were screened for primary LHON mutations. Patients found to have a primary LHON mutation were then tested for 9 proposed secondary LHON mutations. We investigated the association between these mutations and clinical characteristics. RESULTS Primary mtDNA mutations were identified in 68 patients: at np 3460 in 3 (4%) of 68 patients, at np 11,778 in 59 patients (87%), and at np 14,484 in 6 patients (9%). We identified 5 secondary mtDNA mutations (at np 3394, 4216, 7444, 9438 or 13,708) in 10 (15%) of 68 LHON patients and 3 mutations (at np 3394, 4216 or 3708) in 6 (7%) of 90 healthy Japanese individuals. No patient was positive for more than one secondary mutation. The frequency of secondary mutations was similar in the 68 LHON patients and 90 controls. The clinical features of the Japanese patients with any of the 3 primary LHON mutations were similar to those of Caucasian patients, despite different mtDNA backgrounds in these populations. The percentage of patients with familial LHON harboring the 3460 or 14,484 mutations was lower in the Japanese population. CONCLUSIONS Japanese patients with LHON exhibited a very high incidence (87%) of the 11,778 primary mutation. Most of the proposed secondary LHON mutations were rare in the Japanese population and they, except the 7444 mutation, may not influence the clinical features of LHON.


Immunology Today | 1998

APECED: A monogenic autoimmune disease providing new clues to self- tolerance

Pärt Peterson; Kentaro Nagamine; Hamish S. Scott; Maarit Heino; Jun Kudoh; Nobuyoshi Shimizu; Kai Krohn

Abstract The cloning of AIRE , the gene for autoimmune polyendocrinopathy–candidiasis–ectodermal dystrophy (APECED) promises to unravel some of the mysteries of autoimmunity. Here, Part Peterson and colleagues discuss recent studies of AIRE and the implications for disease therapy.


Human Genetics | 1998

Identification and characterization of a novel cyclic nucleotide phosphodiesterase gene (PDE9A) that maps to 21q22.3: alternative splicing of mRNA transcripts, genomic structure and sequence.

Michel Guipponi; Hamish S. Scott; Jun Kudoh; Kazuhiko Kawasaki; Kazunori Shibuya; Ai Shintani; Shuichi Asakawa; Haiming Chen; Maria D. Lalioti; Colette Rossier; Shinsei Minoshima; Nobuyoshi Shimizu

Cyclic nucleotide-specific phosphodiesterases (PDEs) play an essential role in signal transduction by regulating the intracellular concentration of second messengers (cAMP and cGMP). We have identified and made an initial characterization of a full-length cDNA encoding a novel human cyclic nucleotide phosphodiesterase, PDE9A. At least four different mRNA transcripts (PDE9A1, A2, A3, A4) are produced as a result of alternative splicing of 5′ exons, potentially changing the N-terminal amino acid sequences of the encoded proteins. All these predicted proteins would contain a 3′,5′-cyclic nucleotide phosphodiesterase signature motif (Prosite no. PDOC00116). Northern blot analysis revealed several mRNA species of approximately 2.4 kb with varying expression patterns and intensities in most tissues examined, except blood. We have also isolated the mouse homolog of the human PDE9A2 mRNA transcript, pde9A2. The human and mouse isoforms have 93 and 83% sequence identity at the amino acid and nucleotide levels, respectively. PDE9A was mapped to 21q22.3, between TFF1 and D21S360. Comparison of the PDE9A1 cDNA with the genomic sequence from the region revealed that the gene is split into 20 exons that extend over 122 kb. Comparison of the physical map of the region and the genomic sequence further refines the mapping, with D21S113 being derived from intron 15. Several genetic disorders map to 21q22.3, including one form of bipolar affective disorder. Since functional disturbances in intraneuronal signal transmission via second messengers play an important role in the pathophysiology of affective disorders, PDE9A is a strong candidate for such a role by position and function.

Collaboration


Dive into the Jun Kudoh's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hamish S. Scott

Institute of Medical and Veterinary Science

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge