Researcher Database

Researcher Profile and Settings

Master

Affiliation (Master)

  • Faculty of Medicine Research Center for Cooperative Projects

Affiliation (Master)

  • Faculty of Medicine Research Center for Cooperative Projects

researchmap

Profile and Settings

Profile and Settings

  • Name (Japanese)

    Nakamura
  • Name (Kana)

    Hideki
  • Name

    201301040965957050

Achievement

Research Interests

  • 免疫   皮膚病理学   超微形態   細胞・組織   動物   

Research Areas

  • Life sciences / Dermatology

Published Papers

  • Wnt/β-cateninシグナルが表皮角化細胞のヘミデスモソームを制御する
    小住 英之, 野原 拓馬, 藤村 悠, 岩田 浩明, 中村 秀樹, 氏家 英之, 夏賀 健, 渡邉 美佳, Donati Giacomo, 新熊 悟, 築山 忠維
    日本皮膚科学会雑誌 (公社)日本皮膚科学会 132 (12) 2697 - 2697 0021-499X 2022/11
  • Hideyuki Kosumi, Mika Watanabe, Satoru Shinkuma, Takuma Nohara, Yu Fujimura, Tadasuke Tsukiyama, Giacomo Donati, Hiroaki Iwata, Hideki Nakamura, Hideyuki Ujiie, Ken Natsuga
    The Journal of investigative dermatology 142 (6) 1576 - 1586 2022/06 
    Hemidesmosomes (HDs) are adhesion complexes that promote epithelial-stromal attachment in stratified and complex epithelia, including the epidermis. In various biological processes, such as differentiation and migration of epidermal keratinocytes during wound healing or carcinoma invasion, quick assembly and disassembly of HDs are prerequisites. In this study, we show that inhibition of Wnt/β-catenin signaling disturbs HD organization in keratinocytes. Screening with inhibitors identified the depletion of HD components and HD-like structures through Wnt inhibition, but keratinocyte differentiation was not affected. Wnt inhibition significantly diminished plectin and type XVII collagen expression in the basal side of Wnt-inhibited cells and the dermo-epidermal junction of the Wnt-inactive murine basal epidermis. Similar to Wnt inhibition, PLEC-knockout cells or cells with plectin-type XVII collagen binding defects showed type XVII collagen reduction in the basal side of the cells, implying the possible involvement of Wnt/β-catenin signaling in HD assembly. Atypical protein kinase C inhibition ameliorated the phenotypes of Wnt-inhibited cells. These findings show that Wnt/β-catenin signaling regulates the localization of HD components in keratinocytes and that the atypical protein kinase C pathway is involved in Wnt inhibition‒induced HD disarrangement. Our study suggests that the Wnt signaling pathway could be a potential therapeutic target for treating HD-defective diseases, such as epidermolysis bullosa.
  • Ken Natsuga, Yoshikazu Furuta, Shota Takashima, Takuma Nohara, Hsin-Yu Huang, Satoru Shinkuma, Hideki Nakamura, Yousuke Katsuda, Hideaki Higashi, Chao-Kai Hsu, Satoshi Fukushima, Hideyuki Ujiie
    Human mutation 43 (7) 877 - 881 2022/04/21 
    An autosomal recessive disease is caused by biallelic loss-of-function mutations. However, when more than two disease-causing variants are found in a patient's gene, it is challenging to determine which two of the variants are responsible for the disease phenotype. Here, to decipher the pathogenic variants by precise haplotyping, we applied nanopore Cas9-targeted sequencing (nCATS) to three truncation COL7A1 variants detected in a patient with recessive dystrophic epidermolysis bullosa (EB). The distance between the most 5' and 3' variants was approximately 19 kb at the level of genomic DNA. nCATS successfully demonstrated that the most 5' and 3' variants were located in one allele while the variant in between was located in the other allele. Interestingly, the proband's mother, who was phenotypically intact, was heterozygous for the allele that harbored the two truncation variants, which could otherwise be misinterpreted as those of typical recessive dystrophic EB. Our study highlights the usefulness of nCATS as a tool to determine haplotypes of complicated genetic cases. Haplotyping of multiple variants in a gene can determine which variant should be therapeutically targeted when nucleotide-specific gene therapy is applied.
  • 成人栄養障害型表皮水疱症患者を対象としたMuse細胞製品CL2020の国内1/2相試験 52週までの安全性の検討
    藤田 靖幸, 野原 拓馬, 高島 翔太, 夏賀 健, 中村 秀樹, 清水 宏, 足立 太起, 吉田 憲司, 石河 晃, 新熊 悟, 武市 拓也, 秋山 真志, 和田 理
    日本皮膚科学会雑誌 (公社)日本皮膚科学会 132 (3) 510 - 510 0021-499X 2022/03
  • 表皮下水疱は毛包の成長を犠牲にして治癒する
    藤村 悠, 高島 翔太, 中村 秀樹, 小住 英之, 王 禹楠, 眞井 洋輔, 氏家 英之, 岩田 浩明, 西江 渉, 清水 宏, 夏賀 健, 渡邉 美佳, 大野 航太, 小林 康明, 長山 雅晴, Andrea Lauria, Valentina Proserpio, Salvatore Oliviero, Giacomo Donati
    日本皮膚科学会雑誌 (公社)日本皮膚科学会 131 (9) 2071 - 2071 0021-499X 2021/08
  • Yu Fujimura, Mika Watanabe, Kota Ohno, Yasuaki Kobayashi, Shota Takashima, Hideki Nakamura, Hideyuki Kosumi, Yunan Wang, Yosuke Mai, Andrea Lauria, Valentina Proserpio, Hideyuki Ujiie, Hiroaki Iwata, Wataru Nishie, Masaharu Nagayama, Salvatore Oliviero, Giacomo Donati, Hiroshi Shimizu, Ken Natsuga
    EMBO reports 22 (7) 1469-221X 2021/07/05
  • Keisuke Imafuku, Mayumi Kamaguchi, Ken Natsuga, Hideki Nakamura, Hiroshi Shimizu, Hiroaki Iwata
    Cell and tissue research 384 (3) 691 - 702 2021/06 
    Tight junctions (TJs) firmly seal epithelial cells and are key players in the epithelial barrier. TJs consist of several proteins, including those of the transmembrane claudin family and the scaffold zonula occludens (ZO) family. Epithelial tissues are exposed to different conditions: to air in the stratified epithelium of the skin and to liquids in the monolayer of the intestine. The TJs in stratified oral mucosal epithelium have remained insufficiently elucidated in terms of distributions, appearances and barrier functions of TJ proteins in normal buccal mucosa. We investigated these and ZO-1 and claudin-1 were found to be expressed in the top third and in the bottom three quarters of the mucosal epithelium. ZO-1 in the buccal mucosa was found to have an irregular linear appearance. ZO-1 in the buccal mucosa continuously existed in several layers. Electron microscopy revealed the buccal mucosa to have kissing points. In a biotin permeation assay that sought to investigate inside-outside barrier function, the biotin tracer penetrated several ZO-1 layers but did not pass through all the ZO-1 layers. We found that the oral mucosal cell knockdown of TJP1 or CLDN1 resulted in decreases of TER but no significant change in FITC-dextran leakage. Our results suggest that the distribution and appearance of ZO-1 in the buccal mucosa differ from those in the skin. We were unable to prove barrier function in this study but we did show barrier function against small molecules in vivo and against ions in vitro.
  • Y Fujita, T Nohara, S Takashima, K Natsuga, M Adachi, K Yoshida, S Shinkuma, T Takeichi, H Nakamura, O Wada, M Akiyama, A Ishiko, H Shimizu
    Journal of the European Academy of Dermatology and Venereology : JEADV 2021/03/03
  • Matsumura W, Fujita Y, Shinkuma S, Suzuki S, Yokoshiki S, Goto H, Hayashi H, Ono K, Inoie M, Takashima S, Nakayama C, Nomura T, Nakamura H, Abe R, Sato N, Shimizu H
    The Journal of investigative dermatology 139 (10) 2115 - 2124.e11 0022-202X 2019/10 [Refereed][Not invited]
  • Shinkuma S, Nakamura H, Maehara M, Takashima S, Nomura T, Fujita Y, Hasegawa S, Sato-Matsumura KC, Abe R, Shimizu H
    Acta dermato-venereologica 99 (12) 1110 - 1115 0001-5555 2019/09 [Refereed][Not invited]
  • CRISPR/Cas9システムを用いた劣性栄養障害型表皮水疱症(RDEB)の治療法の開発
    高島 翔太, 乃村 俊史, 氏家 英之, 夏賀 健, 岩田 浩明, 中村 秀樹, 清水 宏, 新熊 悟, 阿部 理一郎, 藤田 靖幸
    日本皮膚科学会雑誌 (公社)日本皮膚科学会 129 (9) 1909 - 1909 0021-499X 2019/08
  • Sugai T, Ujiie H, Nakamura H, Kikuchi K, Iwata H, Shimizu H
    The Journal of dermatology 46 (6) e215 - e216 0385-2407 2019/06 [Refereed][Not invited]
  • Takashima S, Shinkuma S, Fujita Y, Nomura T, Ujiie H, Natsuga K, Iwata H, Nakamura H, Vorobyev A, Abe R, Shimizu H
    The Journal of investigative dermatology 139 (8) 1711 - 1721.e4 0022-202X 2019/03 [Refereed][Not invited]
     
    The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system induces site-specific double-strand breaks, which stimulate cellular DNA repair through either the homologous recombination or non-homologous end-joining pathways. The non-homologous end-joining pathway, which is activated more frequently than homologous recombination, is prone to introducing small insertions and/or deletions at the double-strand break site, leading to changes in the reading frame. We hypothesized that the non-homologous end-joining pathway is applicable to genetic diseases caused by a frameshift mutation through restoration of the reading frame. Recessive dystrophic epidermolysis bullosa is a hereditary skin disorder caused by mutations in COL7A1. In this study, we applied gene reframing therapy to a recurrent frameshift mutation, c.5819delC, in COL7A1, which results in a premature termination codon. CRISPR/Cas9 targeting this specific mutation site was delivered to recessive dystrophic epidermolysis bullosa patient fibroblasts. After genotyping a large collection of gene-edited fibroblast clones, we identified a significant number (17/50) of clones in which the frameshift in COL7A1 was restored. The reframed COL7 was functional, as shown by triple-helix formation assay in vitro, and was correctly distributed in the basement membrane zone in mice. Our data suggest that mutation site-specific non-homologous end-joining might be a highly efficient gene therapy for inherited disorders caused by frameshift mutations.
  • Natsuga K, Oiso N, Kurokawa I, Tsubura A, Nakamura H, Maya Y, Nishie W, Kawada A, Shimizu H
    European journal of dermatology : EJD 29 (1) 45 - 48 1167-1122 2019/02 [Refereed][Not invited]
  • Shota Takashima, Satoru Shinkuma, Yasuyuki Fujita, Ken Natsuga, Toshifumi Nomura, Tokimasa Hida, Shuku Ishikawa, Hideki Nakamura, Riichiro Abe, Hiroshi Shimizu
    The Journal of dermatology 45 (9) e260-e261 - e261 2018/09 [Refereed][Not invited]
  • Kosumi H, Nishie W, Sugai T, Toyonaga E, Yoshimoto N, Nakamura H, Horibe R, Kitamura Y, Nakatsumi H, Shimizu H
    Acta dermato-venereologica 98 (4) 454 - 455 0001-5555 2017/12 [Refereed][Not invited]
  • Daisuke Shimbo, Takeo Abumiya, Kota Kurisu, Toshiya Osanai, Hideo Shichinohe, Naoki Nakayama, Ken Kazumata, Hideki Nakamura, Hiroshi Shimuzu, Kiyohiro Houkin
    JOURNAL OF STROKE & CEREBROVASCULAR DISEASES 26 (12) 2994 - 3003 1052-3057 2017/12 [Refereed][Not invited]
     
    Background: The development of cerebral infarction after transient ischemia is attributed to postischemic delayed hypoperfusion in the microvascular region. In the present study, we assessed the microvascular perfusion capacity of infused liposome-encapsulated hemoglobin (LEH) in a therapeutic approach for transient middle cerebral artery occlusion (tMCAO). Methods: Two-hour middle cerebral artery occlusion rats were immediately subjected to intra-arterial infusion of LEH (LEH group) or saline (vehicle group) or no treatment (control group), and then to recanalization. Neurological findings, infarct and edema progression, microvascular endothelial dysfunction, and inflammatory reactions were compared between the 3 groups after 24 hours of reperfusion. Microvascular perfusion in the early phase of reperfusion was evaluated by hemoglobin immunohistochemistry and transmission electron microscopy. Results: The LEH group achieved significantly better results in all items evaluated than the other groups. Hemoglobin immunohistochemistry revealed that the number of hemoglobin-positive microvessels was significantly greater in the LEH group than in the other groups (P < .01), with microvascular perfusion being more likely in narrow microvessels (<= 5 mu m in diameter). An electron microscopic examination revealed that microvessels in the control group were compressed and narrowed by swollen astrocyte end-feet, whereas those in the LEH group had a less deformed appearance and contained LEH particles and erythrocytes. Conclusion: The results of the present study demonstrated that the infusion of LEH reduced infarctions after tMCAO with more hemoglobin-positive and less deformed microvessels at the early phase of reperfusion, suggesting that the superiority of the microvascular perfusion of LEH mediates its neuroprotective effects.
  • Ken Natsuga, Wataru Nishie, Machiko Nishimura, Satoru Shinkuma, Mika Watanabe, Kentaro Izumi, Hideki Nakamura, Yoshiaki Hirako, Hiroshi Shimizu
    HUMAN MUTATION 38 (12) 1666 - 1670 1059-7794 2017/12 [Refereed][Not invited]
     
    Plectin is a linker protein that interacts with intermediate filaments and 4 integrin in hemidesmosomes of the epidermal basement membrane zone (BMZ). Type XVII collagen (COL17) has been suggested as another candidate plectin binding partner in hemidesmosomes. Here, we demonstrate that plectin-COL17 binding helps to maintain epidermal BMZ organization. We identified an epidermolysis bullosa (EB) simplex patient as having markedly diminished expression of plectin and COL17 in skin. The patient is compound heterozygous for sequence variants in the plectin gene (PLEC); one is a truncation and the other is a small in-frame deletion sequence variant. The in-frame deletion is located in the putative COL17-binding domain of plectin and abolishes the plectin-COL17 interaction in vitro. These results imply that disrupted interaction between plectin and COL17 is involved in the development of EB. Our study suggests that protein-protein binding defects may underlie EB in patients with unidentified disease-causing sequence variants.
  • Satoru Shinkuma, Tae Masunaga, Saori Miyawaki, Shota Takashima, Ken Natsuga, Toshifumi Nomura, Yasuyuki Fujita, Hideki Nakamura, Hiroshi Shimizu
    JOURNAL OF DERMATOLOGICAL SCIENCE 88 (1) 139 - 141 0923-1811 2017/10 [Refereed][Not invited]
  • Mika Watanabe, Ken Natsuga, Wataru Nishie, Yasuaki Kobayashi, Giacomo Donati, Shotaro Suzuki, Yu Fujimura, Tadasuke Tsukiyama, Hideyuki Ujiie, Satoru Shinkuma, Hideki Nakamura, Masamoto Murakami, Michitaka Ozaki, Masaharu Nagayama, Fiona M. Watt, Hiroshi Shimizu
    ELIFE 6 2050-084X 2017/07 [Refereed][Not invited]
     
    Type XVII collagen (COL17) is a transmembrane protein located at the epidermal basement membrane zone. COL17 deficiency results in premature hair aging phenotypes and in junctional epidermolysis bullosa. Here, we show that COL17 plays a central role in regulating interfollicular epidermis (IFE) proliferation. Loss of COL17 leads to transient IFE hypertrophy in neonatal mice owing to aberrant Wnt signaling. The replenishment of COL17 in the neonatal epidermis of COL17-null mice reverses the proliferative IFE phenotype and the altered Wnt signaling. Physical aging abolishes membranous COL17 in IFE basal cells because of inactive atypical protein kinase C signaling and also induces epidermal hyperproliferation. The overexpression of human COL17 in aged mouse epidermis suppresses IFE hypertrophy. These findings demonstrate that COL17 governs IFE proliferation of neonatal and aged skin in distinct ways. Our study indicates that COL17 could be an important target of anti-aging strategies in the skin.
  • T. Sugai, S. Shinkuma, K. Inafuku, S. Takashima, T. Nomura, Y. Fujita, H. Nakamura, H. Shimizu
    JOURNAL OF THE EUROPEAN ACADEMY OF DERMATOLOGY AND VENEREOLOGY 31 (5) e251 - e253 0926-9959 2017/05 [Not refereed][Not invited]
  • Shotaro Suzuki, Toshifumi Nomura, Toshinari Miyauchi, Masae Takeda, Hideki Nakamura, Satoru Shinkuma, Yasuyuki Fujita, Masashi Akiyama, Hiroshi Shimizu
    JOURNAL OF INVESTIGATIVE DERMATOLOGY 136 (10) 2093 - 2095 0022-202X 2016/10 [Refereed][Not invited]
  • Kota Kurisu, Takeo Abumiya, Hideki Nakamura, Daisuke Shimbo, Hideo Shichinohe, Naoki Nakayama, Ken Kazumata, Hiroshi Shimizu, Kiyohiro Houkin
    NEUROSURGERY 79 (1) 125 - 134 0148-396X 2016/07 [Refereed][Not invited]
     
    BACKGROUND: Although transarterial regional hypothermia is an attractive alternative to general hypothermia, its efficacy and underlying mechanisms remain unclear. OBJECTIVE: To confirm transarterial regional hypothermia therapeutic effects on ischemia/reperfusion (I/R) injury and to elucidate the mechanisms responsible. METHODS: The therapeutic effects of transarterial regional hypothermia were initially investigated in 2-hour middle cerebral artery occlusion rats regionally infused with 10 degrees C saline (cold saline group) or 37 degrees C saline (warm saline group) and untreated rats (control group) just before the onset of 24 hours of reperfusion. The time course of infarct and edema progression, inflammatory reactions, microvascular morphological changes, and aquaporin-4 (AQP4) expression was analyzed after 0, 2, 6, and 24 hours of reperfusion. RESULTS: Cold saline infusion only lowered brain temperatures for 30 minutes but mediated strong neuroprotective effects with infarct volume reductions of less than one- third. The time-course analysis revealed the following sequence of ischemia/reperfusion injury-related events in the control group: upregulated expression of AQP4 (2 hours); microvascular narrowing resulting from swollen astrocytic end-feet (2-6 hours); infarct and edema progression, blood-brain barrier disruption, and upregulated expression of intracellular adhesion molecule-1 (6-24 hours); and the activation of other inflammatory reactions (24 hours). These sequential events were inhibited in the cold saline group. CONCLUSION: Transarterial regional hypothermia initially inhibited the acute AQP4 surge and then attenuated microvascular narrowing, blood-brain barrier disruption, and activation of other inflammatory reactions, leading to strong neuroprotective effects. More direct and intensive cooling of the endothelium and its surroundings may contribute to these effects.
  • Mayumi Wada, Wataru Nishie, Hideyuki Ujiie, Kentaro Izumi, Hiroaki Iwata, Ken Natsuga, Hideki Nakamura, Yoshimasa Kitagawa, Hiroshi Shimizu
    JOURNAL OF INVESTIGATIVE DERMATOLOGY 136 (5) 938 - 946 0022-202X 2016/05 [Refereed][Not invited]
     
    In bullous pemphigoid, the common autoimmune blistering disorder, IgG autoantibodies target various epitopes on hemidesmosomal transmembrane collagen XVII (COL17)/BP180. Antibodies (Abs) targeting the extracellular noncollagenous 16th A domain of COL17 may be pathogenic; however, the pathogenic roles of Abs targeting non-noncollagenous 16th A regions are poorly understood. In this study using a pathogenic and a nonpathogenic monoclonal antibody (mAb) targeting the noncollagenous 16th A domain (mAb TS39-3) and the C-terminus domain (mAb C17-C1), respectively, we show that endocytosis of immune complexes after binding of Abs to cell surface COL17 is a key phenomenon that induces skin fragility. Passive transfer of IgG1 mouse mAb TS39-3 but not mAb C17-C1 induces dermal-epidermal separation in neonatal human COL17-expressing transgenic mice. Interestingly, mAb C17-C1 strongly binds with the dermal-epidermal junction of the recipient mice skin, suggesting that binding of Abs with COL17 is insufficient to induce skin fragility. In cultured normal human epidermal keratinocytes treated with these mAbs, mAb TS39-3 but not mAb C17-C1 internalizes immune complexes after binding with cell surface COL17 via macropinocytosis, resulting in reduced COL17 expression. This study shows that pathogenicity of Abs targeting COL17 is epitope dependent, which is associated with macropinocytosis-mediated endocytosis of immune complexes and finally results in the depletion of COL17 expression in basal keratinocytes.
  • Machiko Nishimura, Wataru Nishie, Yoshinori Shirafuji, Satoru Shinkuma, Ken Natsuga, Hideki Nakamura, Daisuke Sawamura, Keiji Iwatsuki, Hiroshi Shimizu
    HUMAN MOLECULAR GENETICS 25 (2) 328 - 339 0964-6906 2016/01 [Refereed][Not invited]
     
    In skin, basal keratinocytes in the epidermis are tightly attached to the underlying dermis by the basement membrane (BM). The correct expression of hemidesmosomal and extracellular matrix (ECM) proteins is essential for BM formation, and the null-expression of one molecule may induce blistering diseases associated with immature BM formation in humans. However, little is known about the significance of post-translational processing of hemidesmosomal or ECM proteins in BM formation. Here we show that the C-terminal cleavage of hemidesmosomal transmembrane collagen XVII (COL17) is essential for correct BM formation. The homozygous p.R1303Q mutation in COL17 induces BM duplication and blistering in humans. Although laminin 332, a major ECM protein, interacts with COL17 around p.R1303, the mutation leaves the binding of both molecules unchanged. Instead, the mutation hampers the physiological C-terminal cleavage of COL17 in the ECM. Consequently, non-cleaved COL17 ectodomain remnants induce the aberrant deposition of laminin 332 in the ECM, which is thought to be the major pathogenesis of the BM duplication that results from this mutation. As an example of impaired cleavage of COL17, this study shows that regulated processing of hemidesmosomal proteins is essential for correct BM organization in skin.
  • Wataru Nishie, Ken Natsuga, Hiroaki Iwata, Kentaro Izumi, Hideyuki Ujiie, Ellen Toyonaga, Hiroo Hata, Hideki Nakamura, Hiroshi Shimizu
    AMERICAN JOURNAL OF PATHOLOGY 185 (5) 1361 - 1371 0002-9440 2015/05 [Refereed][Not invited]
     
    Pemphigoid is a common autoimmune blistering disorder in which autoantibodies target transmembrane collagen XVII (COL17), a component of hemidesmosomes in basal keratinocytes. The ectodomain of COL17 can be cleaved from the cell surface within the juxtamembranous extracellular NC16A domain, and, interestingly, certain autoantibodies of pemphigoid patients preferentially react with the shed ectodomain. These findings suggest that COL17 ectodomain shedding generates neoepitopes on the shed form; however, the regulatory mechanism of the shedding in in vivo skin and the pathogenicity of the neoepitope-targeting antibodies still are uncertain. To address these issues, we produced rabbit antibodies specifically reacting with N-terminal cleavage sites of the shed COL17 ectodomain. The antibodies showed that certain amounts of the human COL17 ectodomain are cleaved physiologically at Gln(525) in in vivo skin. In contrast, migrating human keratinocytes cleave COL17 at Leu(524) but not at Gln(525). The passive transfer of antibodies reacting with an N-terminal cleavage site of the mouse COL17 ectodomain into neonatal wild-type mice failed to induce blister formation, even though the antibodies bound to the dermal-epidermal junctions, indicating that cleavage site-specific antibodies have reduced or absent pathogenicity for blister formation. This study shows the ectodomain shedding of COL17 to be a physiological event in in vivo human skin that probably generates nonpathologic epitopes on the cleavage sites.
  • Toyonaga E, Nishie W, Komine M, Murata S, Shinkuma S, Natsuga K, Nakamura H, Ohtsuki M, Shimizu H
    The British journal of dermatology 172 (4) 1141 - 1144 1365-2133 2015/04 [Refereed][Not invited]
     
    Toyonaga E, Nishie W, Komine M, Murata S, Shinkuma S, Natsuga K, Nakamura H, Ohtsuki M, Shimizu H, The British journal of dermatology, 2014
  • Kurisu Kota, Abumiya Takeo, Nakamura Hideki, Shimbo Daisuke, Kazumata Ken, Nakayama Naoki, Shichinohe Hideo, Hokari Masaaki, Osanai Toshiya, Shimizu Hiroshi, Houkin Kiyohiro
    STROKE 46 0039-2499 2015/02 [Refereed][Not invited]
  • Hideyuki Ujiie, Tetsumasa Sasaoka, Kentaro Izumi, Wataru Nishie, Satoru Shinkuma, Ken Natsuga, Hideki Nakamura, Akihiko Shibaki, Hiroshi Shimizu
    JOURNAL OF IMMUNOLOGY 193 (9) 4415 - 4428 0022-1767 2014/11 [Refereed][Not invited]
     
    Complement activation and subsequent recruitment of inflammatory cells at the dermal/epidermal junction are thought to be essential for blister formation in bullous pemphigoid (BP), an autoimmune blistering disease induced by autoantibodies against type XVII collagen (COL17); however, this theory does not fully explain the pathological features of BP. Recently, the involvement of complement-independent pathways has been proposed. To directly address the question of the necessity of the complement activation in blister formation, we generated C3-deficient COL17-humanized mice. First, we show that passive transfer of autoantibodies from BP patients induced blister formation in neonatal C3-deficient COL17-humanized mice without complement activation. By using newly generated human and murine mAbs against the pathogenic noncollagenous 16A domain of COL17 with high (human IgG1, murine IgG2), low (murine IgG1), or no (human IgG4) complement activation abilities, we demonstrate that the deposition of Abs, and not complements, is relevant to the induction of blister formation in neonatal and adult mice. Notably, passive transfer of BP autoantibodies reduced the amount of COL17 in lesional mice skin, as observed in cultured normal human keratinocytes treated with the same Abs. Moreover, the COL17 depletion was associated with a ubiquitin/proteasome pathway. In conclusion, the COL17 depletion induced by BP autoantibodies, and not complement activation, is essential for the blister formation under our experimental system.
  • Natsuga K, Nishie W, Shinkuma S, Nakamura H, Arita K, Yoneda K, Kusaka T, Yanagihara T, Kosaki R, Sago H, Akiyama M, Shimizu H
    Experimental dermatology 20 (1) 74 - 76 0906-6705 2011/01 [Refereed][Not invited]
  • Ken Natsuga, Wataru Nishie, Ken Arita, Satoru Shinkuma, Hideki Nakamura, Shogo Kubota, Sumihisa Imakado, Masashi Akiyama, Hiroshi Shimizu
    JOURNAL OF INVESTIGATIVE DERMATOLOGY 130 (11) 2671 - 2674 0022-202X 2010/11 [Refereed][Not invited]
  • Natsuga K, Nishie W, Shinkuma S, Arita K, Nakamura H, Ohyama M, Osaka H, Kambara T, Hirako Y, Shimizu H
    Human mutation 10 31 E1687 - 98 1059-7794 2010/10 [Refereed][Not invited]
  • Yasuyuki Fujita, Riichiro Abe, Daisuke Inokuma, Mikako Sasaki, Daichi Hoshina, Ken Natsuga, Wataru Nishie, James R. McMillan, Hideki Nakamura, Tadamichi Shimizu, Masashi Akiyama, Daisuke Sawamura, Hiroshi Shimizu
    PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA 107 (32) 14345 - 14350 0027-8424 2010/08 [Refereed][Not invited]
     
    Attempts to treat congenital protein deficiencies using bone marrow-derived cells have been reported. These efforts have been based on the concepts of stem cell plasticity. However, it is considered more difficult to restore structural proteins than to restore secretory enzymes. This study aims to clarify whether bone marrow transplantation (BMT) treatment can rescue epidermolysis bullosa (EB) caused by defects in keratinocyte structural proteins. BMT treatment of adult collagen XVII (Col17) knockout mice induced donor-derived keratinocytes and Col17 expression associated with the recovery of hemidesmosomal structure and better skin manifestations, as well improving the survival rate. Both hematopoietic and mesenchymal stem cells have the potential to produce Col17 in the BMT treatment model. Furthermore, human cord blood CD34(+) cells also differentiated into keratinocytes and expressed human skin component proteins in transplanted immunocompromised (NOD/SCID/gamma(null)(c)) mice. The current conventional BMT techniques have significant potential as a systemic therapeutic approach for the treatment of human EB.
  • Satoru Shinkuma, Masashi Akiyama, Asuka Inoue, Junken Aoki, Ken Natsuga, Toshifumi Nomura, Ken Arita, Riichiro Abe, Kei Ito, Hideki Nakamura, Hideyuki Ujiie, Akihiko Shibaki, Hiraku Suga, Yuichiro Tsunemi, Wataru Nishie, Hiroshi Shimizu
    HUMAN MUTATION 31 (5) 602 - 610 1059-7794 2010/05 [Refereed][Not invited]
     
    Autosomal recessive hypotrichosis (ARH) is characterized by sparse hair on the scalp without other abnormalities. Three genes, DSG4, LIPH, and LPAR6 (P2RY5), have been reported to underlie ARH. We performed a mutation search for the three candidate genes in five independent Japanese ARH families and identified two LIPH mutations: c.736T > A (p.Cys246Ser) in all five families, and c.742C > A (p.His248Asn) in four of the five families. Out of 200 unrelated control alleles, we detected c.736T > A in three alleles and c.742C > A in one allele. Haplotype analysis revealed each of the two mutant alleles is derived from a respective founder. These results suggest the LIPH mutations are prevalent founder mutations for ARH in the Japanese population. LIPH encodes PA-PLA(1)alpha (LIPH), a membrane-associated phosphatidic acid-preferring phospholipase A(1)alpha. Two residues, altered by these mutations, are conserved among PA-PLA(1)alpha of diverse species. Cys(246) forms intramolecular disulfide bonds on the lid domain, a crucial structure for substrate recognition, and His(248) is one amino acid of the catalytic triad. Both p.Cys246Ser- and p.His248Asn-PA-PLA(1)alpha mutants showed complete abolition of hydrolytic activity and had no P2Y5 activation ability. These results suggest defective activation of P2Y5 due to reduced 2-acyl lysophosphatidic acid production by the mutant PA-PLA(1)alpha is involved in the pathogenesis of ARH. Hum Mutat 31:602-610, 2010. (C) 2010 Wiley-Liss, Inc.
  • Ken Natsuga, Wataru Nishie, Masashi Akiyama, Hideki Nakamura, Satoru Shinkuma, James R. McMillan, Akari Nagasaki, Cristina Has, Takeshi Ouchi, Akira Ishiko, Yoshiaki Hirako, Katsushi Owaribe, Daisuke Sawamura, Leena Bruckner-Tuderman, Hiroshi Shimizu
    HUMAN MUTATION 31 (3) 308 - 316 1059-7794 2010/03 [Refereed][Not invited]
     
    Plectin is a cytoskeletal linker protein that has a dumbbell-like structure with a long central rod and N- and C-terminal globular domains. Mutations in the gene encoding plectin (PLEC1) cause two distinct autosomal recessive subtypes of epidermolysis bullosa (EB): EB simplex with muscular dystrophy (EBS-MD), and EB simplex with pyloric atresia (EBS-PA). Here, we demonstrate that normal human fibroblasts express two different plectin isoforms including full-length and rodless forms of plectin. We performed detailed analysis of plectin expression patterns in six EBS-MD and three EBS-PA patients. In EBS-PA, expression of all plectin domains was found to be markedly attenuated or completely lost; in EBS-MD, the expression of the N- and C-terminal domains of plectin remained detectable, although the expression of rod domains was absent or markedly reduced. Our data suggest that loss of the full-length plectin isoform with residual expression of the rodless plectin isoform leads to EBS-MD, and that complete loss or marked attenuation of full-length and rodless plectin expression underlies the more severe EBS-PA phenotype. These results also clearly account for the majority of EBS-MD PLEC1 mutation restriction within the large exon 31 that encodes the plectin rod domain, whereas EBS-PA PLEC1 mutations are generally outside exon 31. Hum Mutat 31:308-316, 2010. (C) 2010 Wiley-Liss, Inc.
  • Gang Wang, Hideyuki Ujiie, Akihiko Shibaki, Wataru Nishie, Yasuki Tateishi, Kazuhiro Kikuchi, Qiang Li, James R. McMillan, Hiroshi Morioka, Daisuke Sawamura, Hideki Nakamura, Hiroshi Shimizu
    AMERICAN JOURNAL OF PATHOLOGY 176 (2) 914 - 925 0002-9440 2010/02 [Refereed][Not invited]
     
    Activation of the complement cascade via the classical pathway is required for the development of tissue injury in many autoantibody-mediated diseases. It therefore makes sense to block the pathological action of autoantibodies by preventing complement activation through inhibition of autoantibody binding to the corresponding pathogenic autoantigen using targeted Fab antibody fragments. To achieve this, we use bullous pemphigoid (BP) as an example of a typical autoimmune disease. Recombinant Fabs against the non-collagenous 16th-A domain of type XVII collagen, the main pathogenic epitope for autoantibodies in BP, were generated from antibody repertoires of BP patients by phage display. Two Fabs, Fab-B4 and Fab-19, showed marked ability to inhibit the binding of BP autoantibodies and subsequent complement activation in vitro. In the in vivo experiments using type XVII collagen humanized BP model mice, these Fabs protected mice against BP autoantibody-induced blistering disease. Thus, the blocking of pathogenic epitopes using engineered Fabs appears to demonstrate efficacy and may lead to disease-specific treatments for antibody-mediated autoimmune diseases. (Am J Pathol 2010, 176:914-925; DOI: 10.2353/ajpath.2010.090744)
  • Hideyuki Ujiie, Akihiko Shibaki, Wataru Nishie, Daisuke Sawamura, Gang Wang, Yasuki Tateishi, Qiang Li, Reine Moriuchi, Hongjiang Qiao, Hideki Nakamura, Masashi Akiyama, Hiroshi Shimizu
    JOURNAL OF IMMUNOLOGY 184 (4) 2166 - 2174 0022-1767 2010/02 [Refereed][Not invited]
     
    Bullous pemphigoid (BP), the most common autoimmune blistering disease, is caused by autoantibodies against type XVII collagen (COL17). To establish an active stable BP animal model that demonstrates the persistent inflammatory skin lesions initiated by the anti-human COL17 Abs, we used COL17-humanized (COL17(m-/-,h+)) mice that we recently produced. First, we generated immunodeficient Rag-2(-/-)/COL17-humanized mice by crossing Rag-2(-/-) mice with COL17-humanized mice. Then, splenocytes from wild-type mice that had been immunized by grafting of human COL17-transgenic mouse skin were transferred into Rag-2(-/-)/COL17-humanized mice. The recipient mice continuously produced anti-human COL17 IgG Abs in vivo and developed blisters and erosions corresponding to clinical, histological, and immunopathological features of BP, although eosinophil infiltration, one of the characteristic histological findings observed in BP patients, was not detected in the recipients. Although the depletion of CD8(+) T cells from the immunized splenocytes was found to produce no effects in the recipients, the depletion of CD4(+) T cells as well as CD45R(+) B cells was found to inhibit the production of anti-human COL17 IgG Abs in the recipients, resulting in no apparent clinical phenotype. Furthermore, we demonstrated that cyclosporin A significantly suppressed the production of anti-human COL17 IgG Abs and prevented the development of the BP phenotype in the treated recipients. Although this model in an immunodeficient mouse does not exactly reproduce the induction mechanism of BP in human patients, this unique experimental system targeting humanized pathogenic Ag allows us to investigate ongoing autoimmune responses to human molecules in experimental animal models. The Journal of Immunology, 2010, 184: 2166-2174.
  • Kei Ito, Daisuke Sawamura, Maki Goto, Hideki Nakamura, Wataru Nishie, Kaori Sakai, Ken Natsuga, Satoru Shinkuma, Akihiko Shibaki, Jouni Uitto, Christopher P. Denton, Osamu Nakajima, Masashi Akiyama, Hiroshi Shimizu
    AMERICAN JOURNAL OF PATHOLOGY 175 (6) 2508 - 2517 0002-9440 2009/12 [Refereed][Not invited]
     
    Recessive dystrophic epidermolysis bullosa (RDEB) is a severe hereditary bullous disease caused by mutations in COL7A1, which encodes type VU collagen (COL7). coral knockout mice (COL7(m-/-)) exhibit a severe RDEB phenotype and die within a few days after birth. Toward developing novel approaches for treating patients with RDEB, we attempted to rescue COL7(m-/-) mice by introducing human COL7A1 cDNA. we first generated transgenic mice that express human COL7A1 cDNA specifically in either epidermal keratinocytes or dermal fibroblasts. We then performed transgenic rescue experiments by crossing these transgenic mice with COL7(m+/-) heterozygous mice. Surprisingly, human COL7 expressed by keratinocytes or by fibroblasts was able to rescue all of the abnormal phenotypic manifestations of the COL7(m-/-) mice, indicating that fibroblasts as well as keratinocytes are potential targets for RDEB gene therapy. Furthermore, we generated transgenic mice with a premature termination codon expressing truncated COL7 protein and performed the same rescue experiments. Notably, the COL7(m-/-) mice rescued with the human COL7A1 allele were able to survive despite demonstrating clinical manifestations very similar to those of human RDEB, indicating that we were able to generate surviving animal models of RDEB with a mutated human COL7A1 gene. This model has great potential for future research into the pathomechanisms of dystrophic epidermolysis bullosa and the development of gene therapies for patients with dystrophic epidermolysis bullosa. (Am J Pathol 2009,175:2508-2517; DOI: 10.2353/ajpath.2009.090347)
  • Daisuke Sawamura, Maki Goto, Kaori Sakai, Hideki Nakamura, James R. McMillan, Masashi Akiyama, Osamu Shirado, Noritaka Oyama, Masataka Satoh, Fumio Kaneko, Toshiaki Takahashi, Hidehiko Konno, Hiroshi Shimizu
    JOURNAL OF INVESTIGATIVE DERMATOLOGY 127 (6) 1537 - 1540 0022-202X 2007/06 [Refereed][Not invited]
  • Wataru Nishie, Daisuke Sawamura, Maki Goto, Kei Ito, Akihiko Shibaki, James R. McMillan, Kaori Sakai, Hideki Nakamura, Edit Olasz, Kim B. Yancey, Masashi Akiyama, Hiroshi Shimizu
    NATURE MEDICINE 13 (3) 378 - 383 1078-8956 2007/03 [Refereed][Not invited]
     
    Transmissibility of characteristic lesions to experimental animals may help us understand the pathomechanism of human autoimmune disease. Here we show that human autoimmune disease can be reproduced using genetically engineered model mice. Bullous pemphigoid ( BP) is the most common serious autoimmune blistering skin disease, with a considerable body of indirect evidence indicating that the underlying autoantigen is collagen XVII (COL17). Passive transfer of human BP autoantibodies into mice does not induce skin lesions, probably because of differences between humans and mice in the amino acid sequence of the COL17 pathogenic epitope. We injected human BP autoantibody into Col17-knockout mice rescued by the human ortholog. This resulted in BP-like skin lesions and a human disease phenotype. Humanization of autoantigens is a new approach to the study of human autoimmune diseases.
  • Abe M, Sawamura D, Goto M, Nakamura H, Nagasaki A, Nomura Y, Kawasaki H, Isogai R, Shimizu H
    J Dermatol Sci 47 165 - 167 2007 [Refereed][Not invited]
  • Yasukawa K, Sawamura D, Goto M, Nakamura H, Shimizu H
    Br J Dermatol 157 662 - 669 2007 [Refereed][Not invited]
  • Inokuma D, Abe R, Fujita Y, Sasaki M, Shibaki A, Nakamura H, McMillan JR, Shimizu T, Shimizu H
    Stem cells (Dayton, Ohio) 12 24 2810 - 2816 1066-5099 2006/12 [Refereed][Not invited]
  • Nakamura H, Sawamura D, Goto M, Nakamura H, Kida M, Ariga T, Sakiyama Y, Tomizawa K, Mitsui H, Tamaki K, Shimizu H
    International journal of molecular medicine 2 18 333 - 337 1107-3756 2006/08 [Refereed][Not invited]
  • Yasukawa K, Sawamura D, Goto M, Nakamura H, Jung SY, Kim SC, Shimizu H
    The British journal of dermatology 2 155 313 - 317 0007-0963 2006/08 [Refereed][Not invited]
  • McMillan JR, Akiyama M, Nakamura H, Shimizu H
    J Histochem Cytochem 54 (1) 109 - 118 0022-1554 2006 [Refereed][Not invited]
  • Sawamura D, Mochitomi Y, Kanzaki T, Nakamura H, Shimizu H
    Br J Dermatol 155 834 - 837 2006 [Refereed][Not invited]
  • Horiguchi Y, Sawamura D, Mori R, Nakamura H, Takahashi K, Shimizu H
    The Journal of investigative dermatology 1 125 83 - 85 0022-202X 2005/07 [Refereed][Not invited]
  • H Mitsui, T Watanabe, M Komine, H Nakamura, H Shimizu, K Tamaki
    JOURNAL OF THE AMERICAN ACADEMY OF DERMATOLOGY 52 (2) 371 - 373 0190-9622 2005/02 [Refereed][Not invited]
  • Nakamura H, Sawamura D, Goto M, McMillan JR, Park S, Kono S, Hasegawa S, Paku S, Nakamura T, Ogiso Y, Shimizu H
    J Mol Diagn 7 28 - 35 2005 [Refereed][Not invited]
  • Sawamura D, Goto M, Yasukawa K, Sato-Matsumura K, Nakamura H, Ito K, Tomita Y, Shimizu H
    J Hum Genet Springer-Verlag Tokyo 50 (10) 543 - 546 1434-5161 2005 [Refereed][Not invited]
     
    Dystrophic EB (DEB) is clinically characterized by mucocutaneous blistering in response to minor trauma, followed by scarring and nail dystrophy, and is caused by mutations in the COL7A1 gene encoding type VII collagen. DEB is inherited in either an autosomal dominant (DDEB) or recessive (RDEB) fashion. DDEB basically results from a glycine substitution mutation within the collagenous domain on one COL7A1 allele, while a combination of mutations such as premature stop codon, missense, splice-site mutations on both alleles causes RDEB. This study performed mutation analysis in 20 distinct Japanese DEB families (16 RDEB and 4 DDEB). The result demonstrated 30 pathogenic COL7A1 mutations with 16 novel mutations, which included 4 missense, 5 nonsense, 1 deletion, 2 insertion, 1 indel, 3 splice-site mutations. We confirmed that Japanese COL7A1 mutations were basically family specific although 3 mutations 5818delC, 6573+1G>C, E2857X were recurrent based on previous reports. Furthermore, Q2827X mutation found in two unrelated families would be regarded as a candidate recurrent Japanese COL7A1 mutation. The study furthers our understanding of both the clinical and allelic heterogeneity displayed in Japanese DEB patients.
  • Tsuji-Abe Y, Akiyama M, Nakamura H, Takizawa Y, Sawamura D, Matsunaga K, Suzumori K, Shimizu H
    Journal of the American Academy of Dermatology 6 51 (6) 1008 - 1011 0190-9622 2004/12 [Refereed][Not invited]
  • Yamagishi S, Abe R, Inagaki Y, Nakamura K, Sugawara H, Inokuma D, Nakamura H, Shimizu T, Takeuchi M, Yoshimura A, Bucala R, Shimizu H, Imaizumi T
    The American journal of pathology 6 165 1865 - 1874 0002-9440 2004/12 [Refereed][Not invited]
  • Goto M, Sato-Matsumura KC, Sawamura D, Yokota K, Nakamura H, Shimizu H
    Journal of dermatological science 3 35 (3) 215 - 220 0923-1811 2004/09 [Refereed][Not invited]
  • Abe R, Shimizu T, Yamagishi S, Shibaki A, Amano S, Inagaki Y, Watanabe H, Sugawara H, Nakamura H, Takeuchi M, Imaizumi T, Shimizu H
    The American journal of pathology 4 164 1225 - 1232 0002-9440 2004/04 [Refereed][Not invited]
  • Abe R, Shimizu T, Sugawara H, Watanabe H, Nakamura H, Choei H, Sasaki N, Yamagishi S, Takeuchi M, Shimizu H
    The Journal of investigative dermatology 2 122 461 - 467 0022-202X 2004/02 [Refereed][Not invited]
  • H Hayashi, T Shimizu, H Nakamura, H Shimizu
    ACTA DERMATO-VENEREOLOGICA 84 (1) 79 - 80 0001-5555 2004 [Refereed][Not invited]
  • Shimizu S, Yasui C, Yasukawa K, Nakamura H, Shimizu H, Tsuchiya K
    The British journal of dermatology 2 149 400 - 404 0007-0963 2003/08 [Refereed][Not invited]
  • H Kawasaki, D Sawamura, F Iwao, T Kikuchi, H Nakamura, S Okubo, T Matsumura, H Shimizu
    BRITISH JOURNAL OF DERMATOLOGY 148 (5) 1047 - 1050 0007-0963 2003/05 [Refereed][Not invited]
     
    We report a 12-year-old boy with nonHallopeau-Siemens recessive dystrophic epidermolysis bullosa (nHS-RDEB) who developed two skin lesions of squamous cell carcinoma (SCC) on the left foot. The incidence of SCC in nHS-RDEB is much lower than in the HS-RDEB subtype. Furthermore, this boy is the youngest among 92 previously described patients with DEB to develop SCC. This study emphasizes the importance of vigilance in monitoring the possible development of SCC in DEB patients regardless of age or clinical severity.
  • Abe R, Shimizu T, Shibaki A, Nakamura H, Watanabe H, Shimizu H
    The American journal of pathology 5 162 1515 - 1520 0002-9440 2003/05 [Refereed][Not invited]
  • Sato-Matsumura KC, Sawamura D, Goto M, Nakamura H, Shimizu H
    Acta Derm Venereol 83 137 - 138 2003 [Refereed][Not invited]
  • K Kimura, KC Sato-Matsumura, H Nakamura, Y Onodera, K Morita, N Enami, T Shougase, T Ohsaki, M Kato, T Takahashi, Y Yamaguchi, H Shimizu
    BRITISH JOURNAL OF DERMATOLOGY 147 (3) 545 - 548 0007-0963 2002/09 [Refereed][Not invited]
     
    Background Fabry disease results from a genetic deficiency of alpha-galactosidase A (GLA) activity. Phenotype-genotype correlations in this condition have not as yet been fully elucidated. Objectives To report a case of a male patient with classical Fabry disease and his mother, a heterozygous female with Fabry disease, showing cardiac involvement, and to identify the underlying GLA gene mutation in this particular phenotype. Patients/methods Genomic DNA was extracted from the patient, his mother and the unaffected family members. Biopsy specimens of skin, heart and kidney were examined using light and electron microscopy. The mutation was identified by polymerase chain reaction and direct sequencing and was confirmed by restriction enzyme fragment length polymorphism. Results The G-->C transversion was identified in codon 97 of the GLA gene and resulted in an A97P amino acid substitution that was a novel pathogenic GLA gene mutation. The male patient who had classical Fabry disease was hemizygous and his mother was heterozygous for this mutation. Conclusions These results indicate that the A97P amino acid substitution in GLA might tend to induce classical Fabry disease.
  • Shimizu T, Nishihira J, Mizue Y, Nakamura H, Abe R, Watanabe H, Ishibashi T, Shimizu H
    Histochemistry and cell biology 3 118 251 - 257 0948-6143 2002/09 [Refereed][Not invited]
  • Shimizu T, Nishihira J, Mizue Y, Nakamura H, Abe R, Watanabe H, Ohkawara A, Shimizu H
    The Journal of investigative dermatology 116 (6) 989 - 990 0022-202X 2001/06 [Refereed][Not invited]
  • T Shimizu, R Abe, H Nakamura, A Ohkawara, M Suzuki, J Nishihira
    BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS 264 (3) 751 - 758 0006-291X 1999/11 [Refereed][Not invited]
     
    Macrophage migration inhibitory factor (MIF) is known to function as a cytokine, hormone, and glucocorticoid-induced immunoregulator. In this study, we reported for the first time that human melanocytes and melanoma cells express MIF mRNA and produce MIF protein. Immunohistochemical analysis demonstrated that MIF was mostly localized in the cytoplasm of melanocytes and G361 cells, a widely available human melanoma cell line. In particular, strong positive staining was observed at the dendrites of these cells. Expression of MIF mRNA and production of MIF protein were much higher in human melanoma cells such as G361, A375, and L32 than in normal cultured melanocytes. To assess the role of MIF overexpression in melanoma cells, G361 cells were transfected with an antisense human MIF plasmid. The results demonstrated that the cell growth rate of the transfected cells was markedly suppressed, suggesting that MIF participates in the mechanism of proliferation of melanoma cells. To further evaluate the function of MIF, we employed the Boyden chamber method to examine the effect on tumor cell migration and found that MIF enhanced the migration of G361 cells in a dose-dependent manner. Furthermore, we administered anti-MIF antibody into tumor (G361 cells in a Millipore chamber)-bearing mice to assess the effect on tumor-associated angiogenesis. The anti-MIF antibody significantly suppressed tumor-induced angiogenesis. Taken together, these results indicated that it is likely that MIF may function as a novel growth factor that stimulates incessant growth and invasion of melanoma concomitant with neovascularization. (C) 1999 Academic Press.

MISC

Research Projects

  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2023/04 -2026/03 
    Author : 中村 秀樹, 柳 輝希, 氏家 英之
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2019/04 -2022/03 
    Author : Nakamura Hideki
     
    The aim of this study was to elucidate the metastatic mechanism of squamous cell carcinoma (SCC) arising in patients with dystrophic epidermolysis bullosa (DEB) caused by mutations in type 7 collagen (COL7) gene. DEB patient model keratinocytes with a comprehensive gene-editing did not develop metastasis when transplanted subcutaneously in mice, but the subcutaneous tumors of DEB patient model showed an overall irregular structure, surrounding fibrosis, and hypervascularization compared to subcutaneous tumors of normal keratinocytes. These results suggest that subcutaneous tumors in DEB patient model induce changes in the surrounding tissue environment via abnormal construction of the basement membrane, and that these changes may contribute to the metastatic mechanism of SCC in DEB patients.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2016/04 -2019/03 
    Author : Nakamura Hideki
     
    Cicatricial alopecia is a type of hair loss characterized by loss of hair follicles and surrounding fibrosis. The aim of this study was to elucidate the mechanism of cicatricial alopecia by analyzing the hair follicles in the blistered skin of mice in detail. A major finding was a delay in hair follicle development/cycling at the re-epithelialized wounds after blistering. At the same time, infiltration of inflammatory cells was poor at the site where several layers of epithelization were completed, and there was no difference compared to the intact site. Taken together, these results indicate that the early stage of cicatricial alopecia involves less inflammation, with the proliferation of epithelial cells required for hair follicle growth directed toward re-epithelialization.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2013/04 -2016/03 
    Author : Nakamura Hideki, NISHIE Wataru, NATSUGA Ken, SHIMIZU Hiroshi
     
    Bullous pemphigoid (BP) is an autoimmune blistering disease. The aim of this study is to address the pathogenic roles of plasmin, a protease present in blister fluids of BP, which is known to cleave COL17. We first identified cleavage sites of COL17 by plasmin, and based on this data, cleavage site-specific antibodies were generated. The antibodies revealed that COL17 is actually cleaved by plasmin in lesional skin of certain numbers of BP patients.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2012/04 -2015/03 
    Author : SHIMIZU Hiroshi, ABE Riichiro, NISHIE Wataru, FUJITA Yasuyuki, AKIYAMA Masashi, KOJIMA Seiji, NAKAMURA Hideki
     
    Epidermolysis bullosa (EB) is a group of genodermatoses that cause blister formations from the congenital abnormality of anchor proteins between the epidermis and the dermis. There have been several strategies for the treatment of EB, and so far, cell therapies are the most promising approach because of the potential of systemic effects. We have proved that stem cell therapies, including bone marrow transplantation, hematopoietic stem cell transplantation, can ameliorate the phenotype and survival prognosis in the junctional EB model mice that lack type XVII collagen (Col17). In this study we explore more efficient approaches of stem cell therapies for EB, including intramedullary transplantation, mesenchymal stromal/stem cell infusion, and investigate factors in association with transdifferentiation from bone marrow-derived stem cells into keratinocytes.
  • Japan Society for the Promotion of Science:Grants-in-Aid for Scientific Research
    Date (from‐to) : 2007 -2008 
    Author : NAKAMURA Hideki, SHIMIZU Hiroshi, NISHIE Wataru
     
    non-Herlitz接合部型表皮水疱症レスキューマウスを詳細に解析し、遺伝子導入治療の客観的評価法を検討した。レスキューマウスに生じる臨床効果の差異は超微細構造学的構造変化と蛋白発現によっても確認することができた。これらの差異が生じる要因として遺伝子導入状態の違いを確認することができたが、この違いを明らかにするためには導入されたgenomic DNAを確認するだけでは不十分であった。臨床効果を分岐する遺伝子導入状態の分岐点を明らかにするためにはrealtime PCRによるmRNAの定量、Fluorescence in-situ Hybridization法による遺伝子導入部位の確認が必要である。


Copyright © MEDIA FUSION Co.,Ltd. All rights reserved.