Junko Morimoto, Ryuichiro Miyazawa, Minoru Matsumoto and Mitsuru Matsumoto : 胸腺上皮細胞を対象とする自己免疫疾患への橋渡し研究の可能性, 北隆館 Precision Medicine 第3巻 第12号, Tokyo, Oct. 2020.
3.
Junko Morimoto, Ryuichiro Miyazawa, Minoru Matsumoto and Mitsuru Matsumoto : 自己免疫疾患(別冊 BIO Clinica 慢性炎症と疾患 通巻24号 第9巻第1巻), HOKURYUKAN, Tokyo, Jul. 2020.
4.
Minoru Matsumoto, Rodrigues M. Pedro, Sousa Laura, Koichi Tsuneyama, Mitsuru Matsumoto and Alves L. Nuno : The Ins and Outs of Thymic Epithelial Cell Differentiation and Function, Springer, Apr. 2019.
Mitsuru Matsumoto : Control of chronic inflammation through elucidation of organ-specific autoimmune disease mechanisms, Springer, Japan, Mar. 2016.
6.
Mitsuru Matsumoto : 内科学, 西村書店, Tokyo, Jul. 2012.
7.
Mitsuru Matsumoto : 遺伝子改変自己免疫モデルーAIRE欠損マウス, エル・アイ・シー, Tokyo, Jun. 2011.
8.
Mitsuru Matsumoto : 生物学事典, Tokyo Kagaku Dozin C., Ltd., Tokyo, Dec. 2010.
Academic Paper (Judged Full Paper):
1.
Minoru Matsumoto, Hideyuki Yoshida, Koichi Tsuneyama, Takeshi Oya and Mitsuru Matsumoto : Revisiting Aire and tissue-restricted antigens at single-cell resolution, Frontiers in Immunology, Vol.14, 1176450, 2023.
(Summary)
The thymus is a highly specialized organ that plays an indispensable role in the establishment of self-tolerance, a process characterized by the "education" of developing T-cells. To provide competent T-cells tolerant to self-antigens, medullary thymic epithelial cells (mTECs) orchestrate negative selection by ectopically expressing a wide range of genes, including various tissue-restricted antigens (TRAs). Notably, recent advancements in the high-throughput single-cell analysis have revealed remarkable heterogeneity in mTECs, giving us important clues for dissecting the mechanisms underlying TRA expression. We overview how recent single-cell studies have furthered our understanding of mTECs, with a focus on the role of Aire in inducing mTEC heterogeneity to encompass TRAs.
Junko Morimoto, Minoru Matsumoto, Takeshi Oya, Koichi Tsuneyama and Mitsuru Matsumoto : Cooperative but Distinct Role of Medullary Thymic Epithelial Cells and Dendritic Cells in the Production of Regulatory T Cells in the Thymus., The Journal of Immunology, 2023.
(Summary)
Regulatory T cells (Tregs) are produced in the thymus to establish self-tolerance, and agonistic stimuli by self-Ags play a pivotal role in this process. Although two types of APCs, medullary thymic epithelial cells (mTECs) and dendritic cells (DCs), are responsible for presenting self-Ags together with costimulatory/cytokine signals, the distinct role of each APC in producing Tregs remains enigmatic. We have approached this issue by depleting the mTECs and DCs using mice expressing diphtheria toxin receptors driven by Aire and CD11c promoters, respectively. Depletion of mTECs showed an effect on Treg production quantitatively and qualitatively more profound than that of DCs followed by the development of distinct organ-specific autoimmune lesions in the hosts. Because self-Ags produced by mTECs are transferable to DCs through a process known as Ag transfer, we monitored the process of Ag transfer using mice expressing GFP from TECs. Although GFP expressed from total TECs was effectively transferred to DCs, GFP expressed from cortical TECs was not, suggesting that mTECs are the predominant source of self-Ags. We also found that GFP expressed not only from mature mTECs but also from immature mTECs was transferred to DCs, suggesting that a broad spectrum of molecules were subjected to Ag transfer during mTEC development. Interestingly, the numbers of recirculating non-Tregs producing IL-2, an important source for Treg expansion in the thymus, were reduced only in the mTEC-depleted mice. These results suggested the cooperative but distinct role of mTECs and DCs in the production of Tregs to avoid autoimmunity.
Minoru Matsumoto, Takuya Ohmura, Yuto Hanibuchi, Mayuko Shimizu, Yasuyo Saijo, Hirohisa Ogawa, Ryuichiro Miyazawa, Junko Morimoto, Koichi Tsuneyama, Mitsuru Matsumoto and Takeshi Oya : AIRE illuminates the feature of medullary thymic epithelial cells in thymic carcinoma., Cancer Medicine, 2023.
(Summary)
Despite the clear distinction between cortical (cTECs) and medullary thymic epithelial cells (mTECs) in physiology, the cell of origin of thymic carcinomas (TCs) and other thymic epithelial tumors remained enigmatic. We addressed this issue by focusing on AIRE, an mTEC-specific transcriptional regulator that is required for immunological self-tolerance. We found that a large proportion of TCs expressed AIRE with typical nuclear dot morphology by immunohistochemistry. AIRE expression in TCs was supported by the RNA-seq data in the TCGA-THYM database. Furthermore, our bioinformatics approach to the recent single-cell RNA-seq data on human thymi has revealed that TCs hold molecular characteristics of multiple mTEC subpopulations. In contrast, TCs lacked the gene signatures for cTECs. We propose that TCs are tumors derived from mTECs.
Ryuichiro Miyazawa, Jun-Ichi Nagao, Ken-Ichi Arita-Morioka, Minoru Matsumoto, Junko Morimoto, Yoshida Masaki, Takeshi Oya, Koichi Tsuneyama, Hedeyuki Yoshida, Yoshihiko Tanaka and Mitsuru Matsumoto : Dispensable Role of Aire in CD11c+ Conventional Dendritic Cells for Antigen Presentation and Shaping the Transcriptome., ImmunoHorizons, Vol.7, No.1, 140-158, 2023.
(Summary)
Aire, the defect of which is responsible for the development of autoimmunity, is predominantly expressed in medullary thymic epithelial cells, and it controls a wide variety of genes, including those of tissue-restricted Ags, for establishing thymic tolerance. Aire is also expressed from APCs in the periphery, called extrathymic Aire-expressing cells (eTACs), and their complementing role to thymic tolerance has been suggested. eTACs are composed of two distinct classes of APCs, conventional dendritic cell (cDC)-type and group 3 innate lymphoid cell (ILC3)-like-type expressing retinoic acid receptor-related orphan receptor γt (RORγt). Although the essential role of Aire in the latter in the Th17-mediated immune response against Candida albicans has been reported, the role of Aire in the cDC-type eTACs for this action has not been examined. Furthermore, the significance of Aire in the production of the transcriptome of the cDC-type eTACs remains unknown. We have approached these issues using a high-fidelity Aire-reporter mouse strain. We found that although the cDC-type eTACs dominated ILC3-like-type eTACs in number and they served as efficient APCs for the immune response against an exogenous Ag as well as for the C. albicans-specific Th17 immune response, loss of Aire in cDC-type eTACs showed no clear effect on these functions. Furthermore, loss of Aire showed no major impact on the transcriptome from cDC-type eTACs. These results suggested that Aire in cDC-type eTACs may not have a cell-intrinsic role in the immune response in contrast to the role of Aire in ILC3-like-type eTACs.
Hitoshi Nishijima, Mizuki Sugita, Natsuka Umezawa, Naoki Kimura, Hirokazu Sasaki, Hiroshi Kawano, Yasuhiko Nishioka, Minoru Matsumoto, Takeshi Oya, Koichi Tsuneyama, Junko Morimoto and Mitsuru Matsumoto : Development of organ-specific autoimmunity by dysregulated Aire expression., Immunology and Cell Biology, Vol.100, No.5, 371-377, 2022.
(Summary)
cells expressing AIRE in the peripheral blood before the treatment but not during the remission phase treated with immunosuppressive drugs. Thus, not only loss of function of AIRE/Aire but also augmented and/or dysregulated expression of AIRE/Aire should be considered for the pathogenesis of organ-specific autoimmunity. We suggest that further analyses should be pursued to establish a novel link between organ-specific autoimmune disease and dysregulated AIRE expression in clinical settings.
Junko Morimoto, Minoru Matsumoto, Ryuichiro Miyazawa, Takeshi Oya, Koichi Tsuneyama and Mitsuru Matsumoto : No Major Impact of Two Homologous Proteins Ly6C1 and Ly6C2 on Immune Homeostasis., ImmunoHorizons, Vol.6, No.3, 202-210, 2022.
(Summary)
Ly6C comprises two homologous components of Ly6C1 and Ly6C2, and the expression of either of the Ly6C molecules defines unique functional subsets of monocytes. Ly6C is also expressed by other immune cell types, including Aire-expressing medullary thymic epithelial cells. Because the role of Ly6C expression in determining the functional subsets remains unclear, we generated mice deficient for both Ly6C1 and Ly6C2 with CRISPR-Cas9mediated deletion. Mice deficient for Ly6C1/Ly6C2 showed no major alterations in the subsets and function of monocyte and other immune cells, including the cells involved in the dextran sulfate sodium saltinduced colitis model. By generating the mice deficient for Ly6C1 alone, we have also investigated the expression pattern of Ly6C1 and Ly6C2 in immune cells. Except for medullary thymic epithelial cells and CD4 single-positive T cells, immune cells predominantly expressed Ly6C2. Thus, despite the importance as a marker with a unique differential expression pattern, the Ly6C molecules have no major impact on determining the functional subsets and maintaining immune homeostasis.
Junko Morimoto, Minoru Matsumoto, Ryuichiro Miyazawa, Hideyuki Yoshida, Koichi Tsuneyama and Mitsuru Matsumoto : Aire suppresses CTLA-4 expression from the thymic stroma to control autoimmunity., Cell Reports, Vol.38, No.7, 110384, 2022.
(Summary)
Impaired production of thymic regulatory T cells (Tregs) is implicated in the development of Aire-dependent autoimmunity. Because Tregs require agonistic T cell receptor stimuli by self-antigens to develop, reduced expression of self-antigens from medullary thymic epithelial cells (mTECs) has been considered to play a major role in the reduced Treg production in Aire deficiency. Here, we show that mTECs abnormally express co-inhibitory receptor CTLA-4 if Aire is non-functional. Upon binding with CD80/CD86 ligands expressed on thymic dendritic cells (DCs), the ectopically expressed CTLA-4 from Aire-deficient mTECs removes the CD80/CD86 ligands from the DCs. This attenuates the ability of DCs to provide co-stimulatory signals and to present self-antigens transferred from mTECs, both of which are required for Treg production. Accordingly, impaired production of Tregs and organ-specific autoimmunity in Aire-deficient mice are rescued by the depletion of CTLA-4 expression from mTECs. Our studies illuminate the significance of mTEC-DC interaction coordinated by Aire for the establishment of thymic tolerance.
Hitoshi Nishijima, Minoru Matsumoto, Junko Morimoto, Kazuyoshi Hosomichi, Nobuko Akiyama, Taishin Akiyama, Takeshi Oya, Koichi Tsuneyama, Hideyuki Yoshida and Mitsuru Matsumoto : Aire Controls Heterogeneity of Medullary Thymic Epithelial Cells for the Expression of Self-Antigens., The Journal of Immunology, Vol.208, No.2, 303-320, 2022.
(Summary)
The deficiency of Aire, a transcriptional regulator whose defect results in the development of autoimmunity, is associated with reduced expression of tissue-restricted self-Ags (TRAs) in medullary thymic epithelial cells (mTECs). Although the mechanisms underlying Aire-dependent expression of TRAs need to be explored, the physical identification of the target(s) of Aire has been hampered by the low and promiscuous expression of TRAs. We have tackled this issue by engineering mice with augmented Aire expression. Integration of the transcriptomic data from Aire-augmented and Aire-deficient mTECs revealed that a large proportion of so-called Aire-dependent genes, including those of TRAs, may not be direct transcriptional targets downstream of Aire. Rather, Aire induces TRA expression indirectly through controlling the heterogeneity of mTECs, as revealed by single-cell analyses. In contrast, Ccl25 emerged as a canonical target of Aire, and we verified this both in vitro and in vivo. Our approach has illuminated the Aire's primary targets while distinguishing them from the secondary targets.
Ferreirinha Pedro, Ribeiro Camila, Junko Morimoto, Landry J M Jonathan, Minoru Matsumoto, Meireles Catarina, White J Andrea, Izumi Ohigashi, Araújo Leonor, Benes Vladimir, Yousuke Takahama, Anderson Graham, Mitsuru Matsumoto and Alves L Nuno : A novel method to identify Post-Aire stages of medullary thymic epithelial cell differentiation, European Journal of Immunology, Vol.51, No.2, 311-318, 2021.
(Summary)
Autoimmune regulator (Aire) medullary thymic epithelial cells (mTECs) play a critical role in tolerance induction. Several studies demonstrated that Aire mTECs differentiate further into Post-Aire cells. Yet, the identification of terminal stages of mTEC maturation depends on unique fate-mapping mouse models. Herein, we resolve this limitation by segmenting the mTEC (MHCII CD80 ) compartment into mTEC (CD24 Sca1 ), mTEC (CD24 Sca1 ), and mTEC (CD24 Sca1 ). While mTEC included mostly Aire-expressing cells, mTEC contained Aire and Aire cells and mTEC were mainly composed of cells lacking Aire. The differential expression pattern of Aire led us to investigate the precursor-product relationship between these subsets. Strikingly, transcriptomic analysis of mTEC , mTEC , and mTEC sequentially mirrored the specific genetic program of Early-, Late- and Post-Aire mTECs. Corroborating their Post-Aire nature, mTEC downregulated the expression of tissue-restricted antigens, acquired traits of differentiated keratinocytes, and were absent in Aire-deficient mice. Collectively, our findings reveal a new and simple blueprint to survey late stages of mTEC differentiation.
Break J Timothy, Oikonomou Vasileios, Dutzan Nicolas, Desai V Jigar, Swidergall Marc, Freiwald Tilo, Chauss Daniel, Harrison J Oliver, Alejo Julie, Williams W Drake, Pittaluga Stefania, Lee R Chyi-Chia, Bouladoux Nicolas, Swamydas Muthulekha, Hoffman W Kevin, Greenwell-Wild Teresa, Bruno M Vincent, Rosen B Lindsey, Lwin Wint, Renteria Andy, Pontejo M Sergio, Shannon P John, Myles A Ian, Olbrich Peter, Ferré M N Elise, Schmitt Monica, Martin Daniel, Core and Computational Biology Genomics, Barber L Daniel, Solis V Norma, Notarangelo D Luigi, Serreze V David, Mitsuru Matsumoto, Hickman D Heather, Murphy M Philip, Anderson S Mark, Lim K Jean, Holland M Steven, Filler G Scott, Afzali Behdad, Belkaid Yasmine, Moutsopoulos M Niki and Lionakis S Michail : Aberrant type 1 immunity drives susceptibility to mucosal fungal infections, Science, Vol.371, No.2021, 1-17, 2021.
(Summary)
Human monogenic disorders have revealed the critical contribution of type 17 responses in mucosal fungal surveillance. We unexpectedly found that in certain settings, enhanced type 1 immunity rather than defective type 17 responses can promote mucosal fungal infection susceptibility. Notably, in mice and humans with deficiency, an autoimmune disease characterized by selective susceptibility to mucosal but not systemic fungal infection, mucosal type 17 responses are intact while type 1 responses are exacerbated. These responses promote aberrant interferon-γ (IFN-γ)- and signal transducer and activator of transcription 1 (STAT1)-dependent epithelial barrier defects as well as mucosal fungal infection susceptibility. Concordantly, genetic and pharmacologic inhibition of IFN-γ or Janus kinase (JAK)-STAT signaling ameliorates mucosal fungal disease. Thus, we identify aberrant T cell-dependent, type 1 mucosal inflammation as a critical tissue-specific pathogenic mechanism that promotes mucosal fungal infection susceptibility in mice and humans.
Minoru Matsumoto, Koichi Tsuneyama, Junko Morimoto, Kazuyoshi Hosomichi, Mitsuru Matsumoto and Hitoshi Nishijima : Tissue-specific autoimmunity controlled by Aire in thymic and peripheral tolerance mechanisms., International Immunology, Vol.32, No.2, 117-131, 2020.
(Summary)
Tissue-specific autoimmune diseases are assumed to arise through malfunction of two checkpoints for immune tolerance: defective elimination of autoreactive T cells in the thymus and activation of these T cells by corresponding autoantigens in the periphery. However, evidence for this model and the outcome of such alterations in each or both of the tolerance mechanisms have not been sufficiently investigated. We studied these issues by expressing human AIRE (huAIRE) as a modifier of tolerance function in NOD mice wherein the defects of thymic and peripheral tolerance together cause type I diabetes (T1D). Additive huAIRE expression in the thymic stroma had no major impact on the production of diabetogenic T cells in the thymus. In contrast, huAIRE expression in peripheral antigen-presenting cells (APCs) rendered the mice resistant to T1D, while maintaining other tissue-specific autoimmune responses and antibody production against an exogenous protein antigen, because of the loss of Xcr1+ dendritic cells, an essential component for activating diabetogenic T cells in the periphery. These results contrast with our recent demonstration that huAIRE expression in both the thymic stroma and peripheral APCs resulted in the paradoxical development of muscle-specific autoimmunity. Our results reveal that tissue-specific autoimmunity is differentially controlled by a combination of thymic function and peripheral tolerance, which can be manipulated by expression of huAIRE/Aire in each or both of the tolerance mechanisms.
Shinya Hatano, Tun Xin, Naoto Noguchi, Yue Dan, Hisakata Yamada, Sun Xun, Mitsuru Matsumoto and Yasunobu Yoshikai : Development of a new monoclonal antibody specific to mouse Vγ6 chain, Life Science Alliance, Vol.2, No.3, 1-12, 2019.
Wang Jianwei, Miho Sekai, Takeshi Matsui, Yosuke Fujii, Mitsuru Matsumoto, Osamu Takeuchi, Nagahiro Minato and Yoko Hamazaki : Hassall's corpuscles with cellular-senescence features maintain IFNα production through neutrophils and pDC activation in the thymus, International Immunology, Vol.31, No.3, 127-139, 2019.
(Summary)
Hassall's corpuscles (HCs) are composed of cornifying, terminally differentiated medullary thymic epithelial cells (mTECs) that are developed under the control of Aire. Here, we demonstrated that HC-mTECs show features of cellular senescence and produce inflammatory cytokines and chemokines including CXCL5, thereby recruiting and activating neutrophils to produce IL-23 in the thymic medulla. We further indicated that thymic plasmacytoid dendritic cells (pDCs) expressing IL-23 receptors constitutively produced Ifna, which plays a role in single positive (SP) cell maturation, in an Il23a-dependent manner. Neutrophil depletion with anti-Ly6G antibody injection resulted in a significant decrease of Ifna expression in the thymic pDCs, suggesting that thymic neutrophil activation underlies the Ifna expression in thymic pDCs in steady state conditions. A New Zealand White mouse strain showing HC hyperplasia exhibited greater numbers and activation of thymic neutrophils and pDCs than B6 mice, whereas Aire-deficient B6 mice with defective HC development and SP thymocyte maturation showed significantly compromised numbers and activation of these cells. These results collectively suggested that HC-mTECs with cell-senescence features initiate a unique cell activation cascade including neutrophils and pDCs leading to the constitutive IFNα expression required for SP T-cell maturation in the thymic medulla.
Junko Morimoto, Yumiko Nishikawa, Takumi Kakimoto, Kohei Furutani, Naoki Kihara, Minoru Matsumoto, Koichi Tsuneyama, Yuko Kozono, Haruo Kozono, Katsuto Hozumi, Kazuyoshi Hosomichi, Hitoshi Nishijima and Mitsuru Matsumoto : Aire Controls in Trans the Production of Medullary Thymic Epithelial Cells Expressing Ly-6C/Ly-6G., The Journal of Immunology, Vol.201, No.11, 3244-3257, 2018.
(Summary)
Medullary thymic epithelial cells (mTECs), which express a wide range of tissue-restricted Ags (TRAs), contribute to the establishment of self-tolerance by eliminating autoreactive T cells and/or inducing regulatory T cells. Aire controls a diverse set of TRAs within Aire-expressing cells by employing various transcriptional pathways. As Aire has a profound effect on transcriptomes of mTECs, including TRAs not only at the single-cell but also the population level, we suspected that Aire (Aire+ mTECs) might control the cellular composition of the thymic microenvironment. In this study, we confirmed that this is indeed the case by identifying a novel mTEC subset expressing Ly-6 family protein whose production was defective in Aire-deficient thymi. Reaggregated thymic organ culture experiments demonstrated that Aire did not induce the expression of Ly-6C/Ly-6G molecules from mTECs as Aire-dependent TRAs in a cell-intrinsic manner. Instead, Aire+ mTECs functioned in trans to maintain Ly-6C/Ly-6G+ mTECs. Thus, Aire not only controls TRA expression transcriptionally within the cell but also controls the overall composition of mTECs in a cell-extrinsic manner, thereby regulating the transcriptome from mTECs on a global scale.
Hitoshi Nishijima, Tatsuya Kajimoto, Yoshiki Matsuoka, Yasuhiro Mouri, Junko Morimoto, Minoru Matsumoto, Hiroshi Kawano, Yasuhiko Nishioka, Hisanori Uehara, Keisuke Izumi, Koichi Tsuneyama, Il-mi Okazaki, Taku Okazaki, Kazuyoshi Hosomichi, Ayako Shiraki, Makoto Shibutani, Kunitoshi Mitsumori and Mitsuru Matsumoto : Paradoxical development of polymyositis-like autoimmunity through augmented expression of autoimmune regulator (AIRE)., Journal of Autoimmunity, Vol.86, 75-92, 2018.
(Summary)
Autoimmunity is prevented by the function of the autoimmune regulator [AIRE (Aire in mice)], which promotes the expression of a wide variety of tissue-restricted antigens (TRAs) from medullary thymic epithelial cells (mTECs) and from a subset of peripheral antigen-presenting cells (APCs). We examined the effect of additive expression of human AIRE (huAIRE) in a model of autoimmune diabetes in NOD mice. Unexpectedly, we observed that mice expressing augmented AIRE/Aire developed muscle-specific autoimmunity associated with incomplete maturation of mTECs together with impaired expression of Aire-dependent TRAs. This led to failure of deletion of autoreactive T cells together with dramatically reduced production of regulatory T cells in the thymus. In peripheral APCs, expression of costimulatory molecules was augmented. We suggest that levels of Aire expression need to be tightly controlled for maintenance of immunological tolerance. Our results also highlight the importance of coordinated action between central tolerance and peripheral tolerance under the common control of Aire.
Yasuhiro Mouri, Yoshihiro Ueda, Tomoyoshi Yamano, Minoru Matsumoto, Koichi Tsuneyama, Tatsuo Kinashi and Mitsuru Matsumoto : Mode of tolerance induction and requirement for Aire are governed by the cell types that express self-antigen and those that present antigen, The Journal of Immunology, Vol.199, No.12, 3959-3971, 2017.
(Summary)
Aire controls the fate of autoreactive thymocytes (i.e., clonal deletion or development into regulatory T cells [Tregs]) through transcriptional control of the expression of tissue-restricted self-antigens (TRAs) from medullary thymic epithelial cells (mTECs) and bone marrow (BM)-derived cells. Although TRAs expressed by mTECs and BM-derived cells are suggested to complement each other to generate a full spectrum of TRAs, little is known about the relative contribution of TRAs from each component for establishment of self-tolerance. Furthermore, the precise role of Aire in specific types of Aire-expressing APCs remains elusive. We have approached these issues by generating two different types of transgenic mouse (Tg) model, which express a prefixed model self-antigen driven by the insulin promoter or the Aire promoter. In the insulin-promoter Tg model, mTECs alone were insufficient for clonal deletion, and BM-derived APCs were required for this action by utilizing Ag transferred from mTECs. In contrast, mTECs alone were able to induce Tregs, although at a much lower efficiency in the absence of BM-derived APCs. Importantly, lack of Aire in mTECs, but not in BM-derived APCs, impaired both clonal deletion and production of Tregs. In the Aire-promoter Tg model, both mTECs and BM-derived APCs could independently induce clonal deletion without Aire, and production of Tregs was impaired by the lack of Aire in mTECs, but not in BM-derived APCs. These results suggest that the fate of autoreactive thymocytes together with the requirement for Aire depend on the cell types that express self-antigens and the types of APCs involved in tolerance induction.
Nobuko Akiyama, Nobukazu Takizawa, Maki Miyauchi, Hiromi Yanai, Ryosuke Tateishi, Miho Shinzawa, Riko Yoshinaga, Masaaki Kurihara, Yosuke Demise, Hisataka Yasuda, Shintaro Yagi, Guoying Wu, Mitsuru Matsumoto, Reiko Sakamoto, Nobuaki Yoshida, Josef M. Penninger, Yasuhiro Kobayashi, Jun-ichiro Inoue and Taishin Akiyama : Identi cation of embryonic precursor cells that di erentiate into thymic epithelial cells expressing autoimmune regulator, The Journal of Experimental Medicine, Vol.213, No.8, 1441-1458, 2016.
(Summary)
Medullary thymic epithelial cells (mTECs) expressing autoimmune regulator (Aire) are critical for preventing the onset of autoimmunity. However, the differentiation program of Aire-expressing mTECs (Aire+ mTECs) is unclear. Here, we describe novel embryonic precursors of Aire+ mTECs. We found the candidate precursors of Aire+ mTECs (pMECs) by monitoring the expression of receptor activator of nuclear factor-κB (RANK), which is required for Aire+ mTEC differentiation. pMECs unex- pectedly expressed cortical TEC molecules in addition to the mTEC markers UEA-1 ligand and RANK and differentiated into mTECs in reaggregation thymic organ culture. Introduction of pMECs in the embryonic thymus permitted long-term mainte- nance of Aire+ mTECs and ef ciently suppressed the onset of autoimmunity induced by Aire+ mTEC de ciency. Mechanistically, pMECs differentiated into Aire+ mTECs by tumor necrosis factor receptor-associated factor 6-dependent RANK signaling. Moreover, nonclassical nuclear factor-κB activation triggered by RANK and lymphotoxin-β receptor signaling promoted pMEC induction from progenitors exhibiting lower RANK expression and higher CD24 expression. Thus, our ndings identi ed two novel stages in the differentiation program of Aire+ mTECs.
Hiroshi Kawano, Hitoshi Nishijima, Junko Morimoto, Fumiko Hirota, Ryoko Morita, Yasuhiro Mouri, Yasuhiko Nishioka and Mitsuru Matsumoto : Aire expression is inherent to most medullary thymic epithelial cells during their differentiation program., The Journal of Immunology, Vol.195, No.11, 5149-5158, 2015.
(Summary)
Aire in medullary thymic epithelial cells (mTECs) plays an important role in the establishment of self-tolerance. Because Aire+ mTECs appear to be a limited subset, they may constitute a unique lineage(s) among mTECs. An alternative possibility is that all mTECs are committed to express Aire in principle, but Aire expression by individual mTECs is conditional. To investigate this issue, we established a novel Aire reporter strain in which endogenous Aire is replaced by the human AIRE-GFP-Flag tag (Aire/hAGF-knockin) fusion gene. The hAGF reporter protein was produced and retained very efficiently within mTECs as authentic Aire nuclear dot protein. Remarkably, snapshot analysis revealed that mTECs expressing hAGF accounted for >95% of mature mTECs, suggesting that Aire expression does not represent a particular mTEC lineage(s). We confirmed this by generating Aire/diphtheria toxin receptor knockin mice in which long-term ablation of Aire+ mTECs by diphtheria toxin treatment resulted in the loss of most mature mTECs beyond the proportion of those apparently expressing Aire. These results suggest that Aire expression is inherent to all mTECs but may occur at particular stage(s) and/or cellular states during their differentiation, thus accounting for the broad impact of Aire on the promiscuous gene expression of mTECs.
Hitoshi Nishijima, Satsuki Kitano, Hitoshi Miyachi, Junko Morimoto, Hiroshi Kawano, Fumiko Hirota, Ryoko Morita, Yasuhiro Mouri, Kiyoshi Masuda, Issei Imoto, Koichi Ikuta and Mitsuru Matsumoto : Ectopic Aire expression in the thymic cortex reveals inherent properties of Aire as a tolerogenic factor within the medulla., The Journal of Immunology, Vol.195, No.10, 4641-4649, 2015.
(Summary)
Cortical thymic epithelial cells (cTECs) and medullary thymic epithelial cells (mTECs) play essential roles in the positive and negative selection of developing thymocytes, respectively. Aire in mTECs plays an essential role in the latter process through expression of broad arrays of tissue-restricted Ags. To determine whether the location of Aire within the medulla is absolutely essential or whether Aire could also function within the cortex for establishment of self-tolerance, we used bacterial artificial chromosome technology to establish a semiknockin strain of NOD-background (β5t/Aire-transgenic) mice expressing Aire under control of the promoter of β5t, a thymoproteasome expressed exclusively in the cortex. Although Aire was expressed in cTECs as typical nuclear dot protein in β5t/Aire-Tg mice, cTECs expressing Aire ectopically did not confer transcriptional expression of either Aire-dependent or Aire-independent tissue-restricted Ag genes. We then crossed β5t/Aire-Tg mice with Aire-deficient NOD mice, generating a strain in which Aire expression was confined to cTECs. Despite the presence of Aire(+) cTECs, these mice succumbed to autoimmunity, as did Aire-deficient NOD mice. The thymic microenvironment harboring Aire(+) cTECs, within which many Aire-activated genes were present, also showed no obvious alteration of positive selection, suggesting that Aire's unique property of generating a self-tolerant T cell repertoire is functional only in mTECs.
Yasuhiro Mouri, Hitoshi Nishijima, Hiroshi Kawano, Fumiko Hirota, Nobuo Sakaguchi, Junko Morimoto and Mitsuru Matsumoto : NF-κB-Inducing Kinase in Thymic Stroma Establishes Central Tolerance by Orchestrating Cross-Talk with Not Only Thymocytes but Also Dendritic Cells., The Journal of Immunology, Vol.193, No.9, 4356-4367, 2014.
(Summary)
Essential roles of NF-κB-inducing kinase (NIK) for the development of medullary thymic epithelial cells (mTECs) and regulatory T cells have been highlighted by studies using a strain of mouse bearing a natural mutation of the NIK gene (aly mice). However, the exact mechanisms underlying the defect in thymic cross-talk leading to the breakdown of self-tolerance in aly mice remain elusive. In this study, we demonstrated that production of regulatory T cells and the final maturation process of positively selected conventional αβ T cells are impaired in aly mice, partly because of a lack of mature mTECs. Of note, numbers of thymic dendritic cells and their expression of costimulatory molecules were also affected in aly mice in a thymic stroma-dependent manner. The results suggest a pivotal role of NIK in the thymic stroma in establishing self-tolerance by orchestrating cross-talk between mTECs and dendritic cells as well as thymocytes. In addition, we showed that negative selection was impaired in aly mice as a result of the stromal defect, which accounts for the development of organ-specific autoimmunity through a lack of normal NIK.
Yumiko Nishikawa, Hitoshi Nishijima, Minoru Matsumoto, Junko Morimoto, Fumiko Hirota, Satoru Takahashi, Hervé Luche, Joerg Hans Fehring, Yasuhiro Mouri and Mitsuru Matsumoto : Temporal lineage tracing of Aire-expressing cells reveals a requirement for Aire in their maturation program., The Journal of Immunology, Vol.192, No.6, 2585-2592, 2014.
(Summary)
Understanding the cellular dynamics of Aire-expressing lineage(s) among medullary thymic epithelial cells (AEL-mTECs) is essential for gaining insight into the roles of Aire in establishment of self-tolerance. In this study, we monitored the maturation program of AEL-mTECs by temporal lineage tracing, in which bacterial artificial chromosome transgenic mice expressing tamoxifen-inducible Cre recombinase under control of the Aire regulatory element were crossed with reporter strains. We estimated that the half-life of AEL-mTECs subsequent to Aire expression was ∼7-8 d, which was much longer than that reported previously, owing to the existence of a post-Aire stage. We found that loss of Aire did not alter the overall lifespan of AEL-mTECs, inconsistent with the previous notion that Aire expression in medullary thymic epithelial cells (mTECs) might result in their apoptosis for efficient cross-presentation of self-antigens expressed by AEL-mTECs. In contrast, Aire was required for the full maturation program of AEL-mTECs, as exemplified by the lack of physiological downregulation of CD80 during the post-Aire stage in Aire-deficient mice, thus accounting for the abnormally increased CD80(high) mTECs seen in such mice. Of interest, increased CD80(high) mTECs in Aire-deficient mice were not mTEC autonomous and were dependent on cross-talk with thymocytes. These results further support the roles of Aire in the differentiation program of AEL-mTECs.
Chikako Odaka, Mathias Hauri-Hohl, Kazuya Takizawa, Yumiko Nishikawa, Masashi Yano, Mitsuru Matsumoto, Richard Boyd and George A. Hollander : TGF-β type II receptor expression in thymic epithelial cells inhibits the development of Hassall's corpuscles in mice, International Immunology, Vol.25, No.11, 633-642, 2013.
(Summary)
Hassall's corpuscles are concentric clusters of keratinized epithelial cells located within the thymic medulla of humans and guinea pigs but are scant in mouse and rat. They are considered to be the terminally differentiated stages of medullary thymic epithelial cells (mTECs) but the mechanisms of their origin are unclear. We have previously deleted the TGF-β type II receptor (TGFβRII) specifically in mouse TECs and reported that these mice have mitigated thymic involution and exhibit earlier reconstitution post-irradiation. In this study, we analyzed the differentiation of mTECs in the TGFβRII-knockout mice. Interestingly, the TGFβRII-knockout mice display enhanced development of Hassall's corpuscles. The expression of Aire, stromal-cell-derived factor 1 and thymic stromal lymphopoietin in the thymi of the TGFβRII-knockout mice was similar to that previously reported for the human thymus. In addition, the putative epithelial progenitor markers MTS20 and MTS24 labeled Hassall's corpuscles in normal mice, but the extent and intensity of this staining were greatly enhanced in Hassall's corpuscles of the TGFβRII-knockout mice. The phosphorylated forms of ERK and JNK were also found in Hassall's corpuscles of the TGFβRII-knockout mice. Taken together, we suggest that TGFβRII-mediated signaling in TECs inhibits their development into Hassall's corpuscles in mice.
Sumiko Yoshida, Ken-ichi Aihara, Yasumasa Ikeda, Yuka Sumitomo-Ueda, Ryoko Uemoto, Kazue Ishikawa, Takayuki Ise, Shusuke Yagi, Takashi Iwase, Yasuhiro Mouri, Matomo Sakari, Takahiro Matsumoto, Ken-ichi Takeyama, Masashi Akaike, Mitsuru Matsumoto, Masataka Sata, Kenneth Walsh, Shigeaki Kato and Toshio Matsumoto : Androgen receptor promotes sex-independent angiogenesis in response to ischemia and is required for activation of vascular endothelial growth factor receptor signaling., Circulation, Vol.128, No.1, 60-71, 2013.
(Summary)
These results document a physiological role of AR in sex-independent angiogenic potency and provide evidence of novel cross-talk between the androgen/AR signaling and VEGF/kinase insert domain protein receptor signaling pathways.
Efficient engulfment of apoptotic cells is critical for maintaining tissue homoeostasis. When phagocytes recognize 'eat me' signals presented on the surface of apoptotic cells, this subsequently induces cytoskeletal rearrangement of phagocytes for the engulfment through Rac1 activation. However, the intracellular signalling cascades that result in Rac1 activation remain largely unknown. Here we show that G-protein-coupled receptor kinase 6 (GRK6) is involved in apoptotic cell clearance. GRK6 cooperates with GIT1 to activate Rac1, which promotes apoptotic engulfment independently from the two known DOCK180/ELMO/Rac1 and GULP1/Rac1 engulfment pathways. As a consequence, GRK6-deficient mice develop an autoimmune disease. GRK6-deficient mice also have increased iron stores in splenic red pulp in which F4/80(+) macrophages are responsible for senescent red blood cell clearance. Our results reveal previously unrecognized roles for GRK6 in regulating apoptotic engulfment and its fundamental importance in immune and iron homoeostasis.
Ayuko Sakane, Ahmed Alamir Mahmoud Abdallah, Kiyoshi Nakano, Kazufumi Honda, Wataru Ikeda, Yumiko Nishikawa, Mitsuru Matsumoto, Natsuki Matsushita, Toshio Kitamura and Takuya Sasaki : Rab13 small G protein and junctional Rab13-binding protein (JRAB) orchestrate actin cytoskeletal organization during epithelial junctional development., The Journal of Biological Chemistry, Vol.287, No.51, 42455-42468, 2012.
(Summary)
During epithelial junctional development, both vesicle transport and reorganization of the actin cytoskeleton must be spatiotemporally regulated. Coordination of these cellular functions is especially important, but the precise mechanism remains elusive. Previously, we identified junctional Rab13-binding protein (JRAB)/molecules interacting with CasL-like 2 (MICAL-L2) as an effector of the Rab13 small G protein, and we found that the Rab13-JRAB system may be involved in the formation of cell-cell adhesions via transport of adhesion molecules. Here, we showed that JRAB interacts with two actin-binding proteins, actinin-1 and -4, and filamentous actin via different domains and regulates actin cross-linking and stabilization through these interactions. During epithelial junctional development, JRAB is prominently enriched in the actin bundle at the free border; subsequently, JRAB undergoes a Rab13-dependent conformational change that is required for maturation of cell-cell adhesion sites. These results suggest that Rab13 and JRAB regulate reorganization of the actin cytoskeleton throughout epithelial junctional development from establishment to maturation of cell-cell adhesion.
(Keyword)
Actin Cytoskeleton / Actinin / Actins / Animals / Cell Line / Cell Membrane / Cytoskeletal Proteins / Epithelial Cells / Epithelium / Humans / Mice / Models, Biological / Protein Binding / Protein Multimerization / Protein Structure, Tertiary / Protein Transport / Tight Junctions / rab GTP-Binding Proteins
Yoshihiro Ueda, Koko Katagiri, Takashi Tomiyama, Kaneki Yasuda, Katsuyoshi Habiro, Tomoya Katakai, Susumu Ikehara, Mitsuru Matsumoto and Tatsuo Kinashi : Mst1 regulates integrin-dependent thymocyte trafficking and antigen recognition in the thymus, Nature Communications, Vol.3, 1098, 2012.
(Summary)
Thymocyte trafficking has an important role in thymic selection. Here we show that the Hippo homologue Mst1 is required for thymocyte migration and antigen recognition by LFA-1 and ICAM-1 within the medulla. Using two-photon imaging of thymic tissues, we found that highly motile mature thymocytes arrest and are activated in the vicinity of rare populations of Aire(+) ICAM-1(hi) medullary thymic epithelia in a negatively selecting environment. Notably, Mst1 deficiency or blocking the cell adhesion molecules LFA-1 and ICAM-1 results in inefficient migration and antigen recognition of CD4(+) thymocytes within the medulla. Consistent with these defects, thymocyte selection is impaired in Mst1(-/-) mice, which display T cell-dependent inflammatory infiltrates in multiple organs and develop autoantibodies. Our results suggest that Mst1 has a key role in regulating thymocyte self-antigen recognition in the medulla.
Yukiko Kuroda, Wataru Sako, Satoshi Goto, Tomoyo Sawada, Daisuke Uchida, Yuishin Izumi, Tetsuya Takahashi, Noriko Kagawa, Masayasu Matsumoto, Mitsuru Matsumoto, Ryosuke Takahashi, Ryuji Kaji and Takao Mitsui : Parkin interacts with Klokin1 for mitochondrial import and maintenance of membrane potential, Human Molecular Genetics, Vol.21, No.5, 991-1003, 2012.
(Summary)
Parkin is a multifunctional protein, including maintaining mitochondrial homeostasis. Recent evidence suggests that Parkin is recruited from the cytoplasm to damaged mitochondria with low membrane potential. We found that intracellular localization of Parkin changed with cellular growth phase. Parkin was preferentially localized in the mitochondria of cultured cells. The mitochondria with large amounts of Parkin showed preserved membrane potentials even during treatment with carbonyl cyanide m-chlorophenylhydrazone. Here we report a novel protein named Klokin 1 that transports Parkin to the mitochondria. Klokin 1 was localized to the mitochondria and enhanced mitochondrial expression of Parkin. Klokin 1 enhanced cell viability in Parkin-silenced cells. Klokin 1 expression was enhanced in the brains of Parkin-deficient mice but not in an autopsied PARK2 brain. Our findings indicate that mitochondrial Parkin prevents mitochondrial depolarization and that Klokin 1 may compensate for Parkin deficiency.
Toll-like receptor-7 (TLR7) and 9, innate immune sensors for microbial RNA or DNA, have been implicated in autoimmunity. Upon activation, TLR7 and 9 are transported from the endoplasmic reticulum (ER) to endolysosomes for nucleic acid sensing by an ER-resident protein, Unc93B1. Little is known, however, about a role for sensor transportation in controlling autoimmunity. TLR9 competes with TLR7 for Unc93B1-dependent trafficking and predominates over TLR7. TLR9 skewing is actively maintained by Unc93B1 and reversed to TLR7 if Unc93B1 loses preferential binding via a D34A mutation. We here demonstrate that mice harboring a D34A mutation showed TLR7-dependent, systemic lethal inflammation. CD4(+) T cells showed marked differentiation toward T helper 1 (Th1) or Th17 cell subsets. B cell depletion abolished T cell differentiation and systemic inflammation. Thus, Unc93B1 controls homeostatic TLR7 activation by balancing TLR9 to TLR7 trafficking.
Yasuhiro Mouri, Masashi Yano, Miho Shinzawa, Yusuke Shimo, Fumiko Hirota, Yumiko Nishikawa, Takuro Nii, Hiroshi Kiyonari, Takaya Abe, Hisanori Uehara, Keisuke Izumi, Koji Tamada, Lieping Chen, Josef M. Penninger, Jun-ichiro Inoue, Taishin Akiyama and Mitsuru Matsumoto : Lymphotoxin signal promotes thymic organogenesis by eliciting RANK expression in the embryonic thymic stroma, The Journal of Immunology, Vol.186, No.9, 5047-5057, 2011.
(Summary)
It has recently become clear that signals mediated by members of the TNFR superfamily, including lymphotoxin-β receptor (LTβR), receptor activator for NF-κB (RANK), and CD40, play essential roles in organizing the integrity of medullary thymic epithelial cells (mTECs) required for the establishment of self-tolerance. However, details of the mechanism responsible for the unique and cooperative action of individual and multiple TNFR superfamily members during mTEC differentiation still remain enigmatic. In this study, we show that the LTβR signal upregulates expression of RANK in the thymic stroma, thereby promoting accessibility to the RANK ligand necessary for mTEC differentiation. Cooperation between the LTβR and RANK signals for optimal mTEC differentiation was underscored by the exaggerated defect of thymic organogenesis observed in mice doubly deficient for these signals. In contrast, we observed little cooperation between the LTβR and CD40 signals. Thus, the LTβR signal exhibits a novel and unique function in promoting RANK activity for mTEC organization, indicating a link between thymic organogenesis mediated by multiple cytokine signals and the control of autoimmunity.
Naoko Wada, Koji Nishifuji, Taketo Yamada, Jun Kudoh, Nobuyoshi Shimizu, Mitsuru Matsumoto, Leena Peltonen, Seiho Nagafuchi and Masayuki Amagai : Aire-dependent thymic expression of desmoglein 3, the autoantigen in pemphigus vulgaris, and its role in T-cell tolerance, The Journal of Investigative Dermatology, Vol.131, No.2, 410-417, 2011.
(Summary)
In the mechanism of thymus-induced central tolerance, the transcription factor Aire has been demonstrated to promote the expression of a wide range of peripheral organ-specific antigens (Ags) in the medullary thymic epithelial cells (mTECs), which serve as self-Ags in negative selection. We examined the expression of desmoglein 3 (Dsg3), the autoantigen in pemphigus vulgaris (PV), in mouse thymus and the involvement of Aire in tolerance to Dsg3. Immunofluorescence and in situ hybridization revealed Dsg3 in single cells or in clusters in ∼3% of mTECs near the cortico-medullary junction of the thymus in C57BL/6 mice. Dsg3-expressing mTECs also expressed some Ags of skin-unrelated peripheral organs simultaneously. In contrast, Dsg3-positive mTECs were not detected in the Aire(-/-) thymus. Adoptive transfer of splenocytes from Aire(-/-) mice immunized with Dsg3 did not induce anti-Dsg3 IgG production or PV phenotype in Rag2(-/-) recipient mice. However, Aire(-/-) CD4(+) T cells, but not Aire(+/+) CD4(+) T cells, induced low levels of anti-Dsg3 IgG production when transferred with Dsg3(-/-) B cells. These findings indicate that Aire has an important role in Dsg3 expression as well as in selection of T cells that help B cells to produce anti-Dsg3 IgG in thymus.
Yumiko Nishikawa, Fumiko Hirota, Masashi Yano, Kitajima Hiroyuki, Miyazaki Jun-ichi, Kawamoto Hiroshi, Yasuhiro Mouri and Mitsuru Matsumoto : Biphasic Aire expression in early embryos and in medullary thymic epithelial cells before end-stage terminal differentiation, The Journal of Experimental Medicine, Vol.207, No.5, 963-971, 2010.
(Summary)
The roles of autoimmune regulator (Aire)-expressing medullary thymic epithelial cells (mTECs) in the organization of the thymic microenvironment for establishing self-tolerance are enigmatic. We sought to monitor the production and maintenance of Aire-expressing mTECs by a fate-mapping strategy in which bacterial artificial chromosome transgenic (Tg) mice expressing Cre recombinase under the control of the Aire regulatory element were crossed with a GFP reporter strain. We found that, in addition to its well recognized expression within mature mTECs, Aire was expressed in the early embryo before emergence of the three germ cell layers. This observation may help to explain the development of ectodermal dystrophy often seen in patients with AIRE deficiency. With the use of one Tg line in which Cre recombinase expression was confined to mTECs, we found that Aire(+)CD80(high) mTECs further progressed to an Aire(-)CD80(intermediate) stage, suggesting that Aire expression is not constitutive from after its induction until cell death but instead is down-regulated at the beginning of terminal differentiation. We also demonstrated that many mTECs of Aire-expressing lineage are in close contact with thymic dendritic cells. This close proximity may contribute to transfer of tissue-restricted self-antigens expressed by mTECs to professional antigen-presenting cells.
Yuka Tomoe, Hiroko Segawa, Kazuyo Shiozawa, Ichiro Kaneko, Rieko Tominaga, Etsuyo Hanabusa, Fumito Aranami, Junya Furutani, Shoji Kuwahara, Sawako Tatsumi, Mitsuru Matsumoto, Mikiko Ito and Ken-ichi Miyamoto : Phosphaturic action of fibroblast growth factor 23 in Npt2 null mice, American Journal of Physiology, Renal Physiology, Vol.298, No.6, F1341-F1350, 2010.
(Summary)
In the present study, we evaluated the roles of type II and type III sodium-dependent P(i) cotransporters in fibroblast growth factor 23 (FGF23) activity by administering a vector encoding FGF23 with the R179Q mutation (FGF23M) to wild-type (WT) mice, Npt2a knockout (KO) mice, Npt2c KO mice, and Npt2a(-/-)Npt2c(-/-) mice (DKO mice). In Npt2a KO mice, FGF23M induced severe hypophosphatemia and markedly decreased the levels of Npt2c, type III Na-dependent P(i) transporter (PiT2) protein, and renal Na/P(i) transport activity. In contrast, in Npt2c KO mice, FGF23M decreased plasma phosphate levels comparable to those in FGF23M-injected WT mice. In DKO mice with severe hypophosphatemia, FGF23M administration did not induce an additional increase in urinary phosphate excretion. FGF23 administration significantly decreased intestinal Npt2b protein levels in WT mice but had no effect in Npt2a, Npt2c, and DKO mice, despite marked suppression of plasma 1,25(OH)(2)D(3) levels in all the mutant mice. The main findings were as follow: 1) FGF23-dependent phosphaturic activity in Npt2a KO mice is dependent on renal Npt2c and PiT-2 protein; 2) in DKO mice, renal P(i) reabsorption is not further decreased by FGF23M, but renal vitamin D synthesis is suppressed; and 3) downregulation of intestinal Npt2b may be mediated by a factor(s) other than 1,25(OH)(2)D(3). These findings suggest that Npt2a, Npt2c, and PiT-2 are necessary for the phosphaturic activity of FGF23. Thus complementary regulation of Npt2 family proteins may be involved in systemic P(i) homeostasis.
(Keyword)
Animals / Calcitriol / calcium / Fibroblast Growth Factors / Gene Transfer Techniques / Humans / hypophosphatemia / Hypophosphatemia, Familial / Injections, Intravenous / Mice / Mice, Inbred C57BL / Mice, Knockout / Mutation / Phosphates / Sodium-Phosphate Cotransporter Proteins, Type III / Sodium-Phosphate Cotransporter Proteins, Type IIa / Sodium-Phosphate Cotransporter Proteins, Type IIb / Sodium-Phosphate Cotransporter Proteins, Type IIc
Kimi Yamakoshi, Akiko Takahashi, Fumiko Hirota, Rika Nakayama, Naozumi Ishimaru, Yoshiaki Kubo, J. David Mann, Masako Ohmura, Atsushi Hirao, Hideyuki Saya, Seiji Arase, Yoshio Hayashi, Kazuki Nakao, Mitsuru Matsumoto, Naoko Ohtani and Eiji Hara : Real-time in vivo imaging of p16Ink4a reveals cross-talk with p53, The Journal of Cell Biology, Vol.186, No.3, 393-407, 2009.
(Summary)
Expression of the p16(Ink4a) tumor suppressor gene, a sensor of oncogenic stress, is up-regulated by a variety of potentially oncogenic stimuli in cultured primary cells. However, because p16(Ink4a) expression is also induced by tissue culture stress, physiological mechanisms regulating p16(Ink4a) expression remain unclear. To eliminate any potential problems arising from tissue culture-imposed stress, we used bioluminescence imaging for noninvasive and real-time analysis of p16(Ink4a) expression under various physiological conditions in living mice. In this study, we show that oncogenic insults such as ras activation provoke epigenetic derepression of p16(Ink4a) expression through reduction of DNMT1 (DNA methyl transferase 1) levels as a DNA damage response in vivo. This pathway is accelerated in the absence of p53, indicating that p53 normally holds the p16(Ink4a) response in check. These results unveil a backup tumor suppressor role for p16(Ink4a) in the event of p53 inactivation, expanding our understanding of how p16(Ink4a) expression is regulated in vivo.
Hiroko Segawa, Akemi Onitsuka, Junya Furutani, Ichiro Kaneko, Fumito Aranami, Natsuki Matsumoto, Yuka Tomoe, Masashi Kuwahata, Mikiko Ito, Mitsuru Matsumoto, Minqi Li, Norio Amizuka and Ken-ichi Miyamoto : Npt2a and Npt2c in mice play distinct and synergistic roles in inorganic phosphate metabolism and skeletal development, American Journal of Physiology, Renal Physiology, Vol.297, No.3, F671-678, 2009.
(Summary)
Hereditary hypophosphatemic rickets with hypercalciuria (HHRH) is a rare autosomal recessively inherited disorder, characterized by hypophosphatemia, short stature, rickets and/or osteomalacia, and secondary absorptive hypercalciuria. HHRH is caused by a defect in the sodium-dependent phosphate transporter (NaPi-IIc/Npt2c/NPT2c), which was thought to have only a minor role in renal phosphate (P(i)) reabsorption in adult mice. In fact, mice that are null for Npt2c (Npt2c(-/-)) show no evidence for renal phosphate wasting when maintained on a diet with a normal phosphate content. To obtain insights and the relative importance of Npt2a and Npt2c, we now studied Npt2a(-/-)Npt2c(+/+), Npt2a(+/-)Npt2c(-/-), and Npt2a(-/-)Npt2c(-/-) double-knockout (DKO). DKO mice exhibited severe hypophosphatemia, hypercalciuria, and rickets. These findings are different from those in Npt2a KO mice that show only a mild phosphate and bone phenotype that improve over time and from the findings in Npt2c KO mice that show no apparent abnormality in the regulation of phosphate homeostasis. Because of the nonredundant roles of Npt2a and Npt2c, DKO animals showed a more pronounced reduction in P(i) transport activity in the brush-border membrane of renal tubular cells than that in the mice with the single-gene ablations. A high-P(i) diet after weaning rescued plasma phosphate levels and the bone phenotype in DKO mice. Our findings thus showed in mice that Npt2a and Npt2c have independent roles in the regulation of plasma P(i) and bone mineralization.
Hiroko Segawa, Akemi Onitsuka, Masashi Kuwahata, Etsuyo Hanabusa, Junya Furutani, Ichiro Kaneko, Yuka Tomoe, Fumito Aranami, Natsuki Matsumoto, Mikiko Ito, Mitsuru Matsumoto, Minqi Li, Norio Amizuka and Ken-ichi Miyamoto : Type IIc sodium dependent phosphate transporter regulates calcium metabolism, Journal of the American Society of Nephrology, Vol.20, No.1, 104-113, 2009.
(Summary)
Primary renal inorganic phosphate (Pi) wasting leads to hypophosphatemia, which is associated with skeletal mineralization defects. In humans, mutations in the gene encoding the type IIc sodium-dependent phosphate transporter lead to hereditary hypophophatemic rickets with hypercalciuria, but whether Pi wasting directly causes the bone disorder is unknown. Here, we generated Npt2c-null mice to define the contribution of Npt2c to Pi homeostasis and to bone abnormalities. Homozygous mutants (Npt2c(-/-)) exhibited hypercalcemia, hypercalciuria, and elevated plasma 1,25-dihydroxyvitamin D(3) levels, but they did not develop hypophosphatemia, hyperphosphaturia, renal calcification, rickets, or osteomalacia. The increased levels of 1,25-dihydroxyvitamin D(3) in Npt2c(-/-) mice compared with age-matched Npt2c(+/+) mice may be the result of reduced catabolism, because we observed significantly reduced expression of renal 25-hydroxyvitamin D-24-hydroxylase mRNA but no change in 1alpha-hydroxylase mRNA levels. Enhanced intestinal absorption of calcium (Ca) contributed to the hypercalcemia and increased urinary Ca excretion. Furthermore, plasma levels of the phosphaturic protein fibroblast growth factor 23 were significantly decreased in Npt2c(-/-) mice. Sodium-dependent Pi co-transport at the renal brush border membrane, however, was not different among Npt2c(+/+), Npt2c(+/-), and Npt2c(-/-) mice. In summary, these data suggest that Npt2c maintains normal Ca metabolism, in part by modulating the vitamin D/fibroblast growth factor 23 axis.
(Keyword)
Animals / Biological Transport / Bone and Bones / Calcium / Calcium Channels / Calcium-Binding Protein, Vitamin D-Dependent / Fasting / Female / Fibroblast Growth Factors / Male / Mice / Mice, Inbred C57BL / Phosphates / RNA, Messenger / Sodium-Phosphate Cotransporter Proteins, Type IIc / TRPV Cation Channels
Masashi Yano, Noriyuki Kuroda, Hongwei Han, Makiko Meguro-Horike, Yumiko Nishikawa, Hiroshi Kiyonari, Kentaro Maemura, Yuchio Yanagawa, Kunihiko Obata, Satoru Takahashi, Tomokatsu Ikawa, Rumi Satoh, Hiroshi Kawamoto, Yasuhiro Mouri and Mitsuru Matsumoto : Aire controls the differentiation program of thymic epithelial cells in the medulla for the establishment of self-tolerance., The Journal of Experimental Medicine, Vol.205, No.12, 2827-2838, 2008.
(Summary)
The roles of autoimmune regulator (Aire) in the expression of the diverse arrays of tissue-restricted antigen (TRA) genes from thymic epithelial cells in the medulla (medullary thymic epithelial cells [mTECs]) and in organization of the thymic microenvironment are enigmatic. We approached this issue by creating a mouse strain in which the coding sequence of green fluorescent protein (GFP) was inserted into the Aire locus in a manner allowing concomitant disruption of functional Aire protein expression. We found that Aire(+) (i.e., GFP(+)) mTECs were the major cell types responsible for the expression of Aire-dependent TRA genes such as insulin 2 and salivary protein 1, whereas Aire-independent TRA genes such as C-reactive protein and glutamate decarboxylase 67 were expressed from both Aire(+) and Aire(-) mTECs. Remarkably, absence of Aire from mTECs caused morphological changes together with altered distribution of mTECs committed to Aire expression. Furthermore, we found that the numbers of mTECs that express involucrin, a marker for terminal epidermal differentiation, were reduced in Aire-deficient mouse thymus, which was associated with nearly an absence of Hassall's corpuscle-like structures in the medulla. Our results suggest that Aire controls the differentiation program of mTECs, thereby organizing the global mTEC integrity that enables TRA expression from terminally differentiated mTECs in the thymic microenvironment.
Taishin Akiyama, Yusuke Shimo, Hiromi Yanai, Junwen Qin, Daisuke Ohshima, Yuya Maruyama, Yukiko Asaumi, Juli Kitazawa, Hiroshi Takayanagi, Josef Penninger, Mitsuru Matsumoto, Takeshi Nitta, Yousuke Takahama and Jun-ichiro Inoue : The tumor necrosis factor family receptors RANK and CD40 cooperatively establish the thymic medullary microenvironment and self-tolerance., Immunity, Vol.29, No.3, 423-437, 2008.
(Summary)
Medullary thymic epithelial cells (mTECs) establish T cell self-tolerance through the expression of autoimmune regulator (Aire) and peripheral tissue-specific self-antigens. However, signals underlying mTEC development remain largely unclear. Here, we demonstrate crucial regulation of mTEC development by receptor activator of NF-kappaB (RANK) and CD40 signals. Whereas only RANK signaling was essential for mTEC development during embryogenesis, in postnatal mice, cooperation between CD40 and RANK signals was required for mTEC development to successfully establish the medullary microenvironment. Ligation of RANK or CD40 on fetal thymic stroma in vitro induced mTEC development in a tumor necrosis factor-associated factor 6 (TRAF6)-, NF-kappaB inducing kinase (NIK)-, and IkappaB kinase beta (IKKbeta)-dependent manner. These results show that developmental-stage-dependent cooperation between RANK and CD40 promotes mTEC development, thereby establishing self-tolerance.
Yu Hikosaka, Takeshi Nitta, Izumi Ohigashi, Kouta Yano, Naozumi Ishimaru, Yoshio Hayashi, Mitsuru Matsumoto, Koichi Matsuo, Josef M Penninger, Hiroshi Takayanagi, Yoshifumi Yokota, Hisakata Yamada, Yasunobu Yoshikai, Jun-ichiro Inoue, Taishin Akiyama and Yousuke Takahama : The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator., Immunity, Vol.29, No.3, 438-450, 2008.
(Summary)
The thymic medulla provides a microenvironment where medullary thymic epithelial cells (mTECs) express autoimmune regulator and diverse tissue-restricted genes, contributing to launching self-tolerance. Positive selection is essential for thymic medulla formation via a previously unknown mechanism. Here we show that the cytokine RANK ligand (RANKL) was produced by positively selected thymocytes and regulated the cellularity of mTEC by interacting with RANK and osteoprotegerin. Forced expression of RANKL restored thymic medulla in mice lacking positive selection, whereas RANKL perturbation impaired medulla formation. These results indicate that RANKL produced by positively selected thymocytes is responsible for fostering thymic medulla formation, thereby establishing central tolerance.
Mitsuru Matsumoto, Yiqing Zhou, Shinji Matsuo, Hideki Nakanishi, Kenji Hirose, Hajimu Oura, Seiji Arase, Akemi Ishida-Yamamoto, Yoshimi Bando, Keisuke Izumi, Hiroshi Kiyonari, Naoko Oshima, Rika Nakayama, Akemi Matsushima, Fumiko Hirota, Yasuhiro Mouri, Noriyuki Kuroda, Shigetoshi Sano and David D. Chaplin : Targeted deletion of the murine corneodesmosin gene delineates its essential role in skin and hair physiology, Proceedings of the National Academy of Sciences of the United States of America, Vol.105, No.18, 6720-6724, 2008.
(Summary)
Controlled proteolytic degradation of specialized junctional structures, corneodesmosomes, by epidermal proteases is an essential process for physiological desquamation of the skin. Corneodesmosin (CDSN) is an extracellular component of corneodesmosomes and, although considerable debate still exists, genetic studies have suggested that the CDSN gene in the major psoriasis-susceptibility locus (PSORS1) may be responsible for susceptibility to psoriasis, a human skin disorder characterized by excessive growth and aberrant differentiation of keratinocytes. CDSN is also expressed in the inner root sheath of hair follicles, and a heterozygous nonsense mutation of the CDSN gene in humans is associated with scalp-specific hair loss of poorly defined etiology. Here, we have investigated the pathogenetic roles of CDSN loss of function in the development of skin diseases by generating a mouse strain with targeted deletion of the Cdsn gene. Cdsn-deficient mouse skin showed detachment of the stratum corneum from the underlying granular layer and/or detachment within the upper granular layers due to the disrupted integrity of the corneodesmosomes. When grafted onto immunodeficient mice, Cdsn-deficient skin showed rapid hair loss together with epidermal abnormalities resembling psoriasis. These results underscore the essential roles of CDSN in hair physiology and suggest functional relevance of CDSN gene polymorphisms to psoriasis susceptibility.
Masashi Nakatani, Yuka Takehara, Hiromu Sugino, Mitsuru Matsumoto, Osamu Hashimoto, Yoshihisa Hasegawa, Tatsuya Murakami, Akiyoshi Uezumi, Shin'ichi Takeda, Sumihare Noji, Yoshihide Sunada and Kunihiro Tsuchida : Transgenic expression of a myostatin inhibitor derived from follistatin increases skeletal muscle mass and ameliorates dystrophic pathology in mdx mice., The FASEB journal, Vol.22, No.2, 477-487, 2008.
(Summary)
Myostatin is a potent negative regulator of skeletal muscle growth. Therefore, myostatin inhibition offers a novel therapeutic strategy for muscular dystrophy by restoring skeletal muscle mass and suppressing the progression of muscle degeneration. The known myostatin inhibitors include myostatin propeptide, follistatin, follistatin-related proteins, and myostatin antibodies. Although follistatin shows potent myostatin-inhibiting activities, it also acts as an efficient inhibitor of activins. Because activins are involved in multiple functions in various organs, their blockade by follistatin would affect multiple tissues other than skeletal muscles. In the present study, we report the characterization of a myostatin inhibitor derived from follistatin, which does not affect activin signaling. The dissociation constants (K(d)) of follistatin to activin and myostatin are 1.72 nM and 12.3 nM, respectively. By contrast, the dissociation constants (K(d)) of a follistatin-derived myostatin inhibitor, designated FS I-I, to activin and myostatin are 64.3 microM and 46.8 nM, respectively. Transgenic mice expressing FS I-I, under the control of a skeletal muscle-specific promoter showed increased skeletal muscle mass and strength. Hyperplasia and hypertrophy were both observed. We crossed FS I-I transgenic mice with mdx mice, a model for Duchenne muscular dystrophy. Notably, the skeletal muscles in the mdx/FS I-I mice showed enlargement and reduced cell infiltration. Muscle strength is also recovered in the mdx/FS I-I mice. These results indicate that myostatin blockade by FS I-I has a therapeutic potential for muscular dystrophy.
Junwen Qin, Hiroyasu Konno, Daisuke Ohshima, Hiromi Yanai, Hidehiko Motegi, Yusuke Shimo, Fumiko Hirota, Mitsuru Matsumoto, Satoshi Takaki, Jun-ichiro Inoue and Taishin Akiyama : Developmental stage-dependent collaboration between the TNF receptor-associated factor 6 and lymphotoxin pathways for B cell follicle organization in secondary lymphoid organs, The Journal of Immunology, Vol.179, No.10, 6799-6807, 2007.
(Summary)
Signal transduction pathways regulating NF-kappaB activation essential for microenvironment formation in secondary lymphoid organs remain to be determined. We investigated the effect of a deficiency of TNFR-associated factor 6 (TRAF6), which activates the classical NF-kappaB pathway, in splenic microenvironment formation. Two-week-old TRAF6-deficient mice showed severe defects in B cell follicle and marginal zone formation, similar to mutant mice defective in lymphotoxin (Lt) beta receptor (LtbetaR) signal induction of nonclassical NF-kappaB activation. However, analysis revealed a TRAF6 role in architecture formation distinct from its role in the early neonatal Lt signaling pathway. LtbetaR signal was essential for primary B cell cluster formation with initial differentiation of follicular dendritic cells (FDCs) in neonatal mice. In contrast, TRAF6 was dispensable for progression to this stage but was required for converting B cell clusters to B cell follicles and maintaining FDCs through to later stages. Fetal liver transfer experiments suggested that TRAF6 in radiation-resistant cells is responsible for follicle formation. Despite FDC-specific surface marker expression, FDCs in neonatal TRAF6-deficient mice had lost the capability to express CXCL13. These data suggest that developmentally regulated activation of TRAF6 in FDCs is required for inducing CXCL13 expression to maintain B cell follicles.
Naoko Ohtani, Yuko Imamura, Kimi Yamakoshi, Fumiko Hirota, Rika Nakayama, Yoshiaki Kubo, Naozumi Ishimaru, Akiko Takahashi, Atsushi Hirao, Takatsune Shimizu, David J. Mann, Hideyuki Saya, Yoshio Hayashi, Seiji Arase, Mitsuru Matsumoto, Nakao Kazuki and Eiji Hara : Visualizing the dynamics of p21 (Wafl/Cip1)cyclin-dependent kinase inhibitor expression in living animals., Proceedings of the National Academy of Sciences of the United States of America, Vol.104, No.38, 15034-15039, 2007.
(Summary)
Although the role of p21(Waf1/Cip1) gene expression is well documented in various cell culture studies, its in vivo roles are poorly understood. To gain further insight into the role of p21(Waf1/Cip1) gene expression in vivo, we attempted to visualize the dynamics of p21(Waf1/Cip1) gene expression in living animals. In this study, we established a transgenic mice line (p21-p-luc) expressing the firefly luciferase under the control of the p21(Waf1/Cip1) gene promoter. In conjunction with a noninvasive bioluminescent imaging technique, p21-p-luc mice enabled us to monitor the endogenous p21(Waf1/Cip1) gene expression in vivo. By monitoring and quantifying the p21(Waf1/Cip1) gene expression repeatedly in the same mouse throughout its entire lifespan, we were able to unveil the dynamics of p21(Waf1/Cip1) gene expression in the aging process. We also applied this system to chemically induced skin carcinogenesis and found that the levels of p21(Waf1/Cip1) gene expression rise dramatically in benign skin papillomas, suggesting that p21(Waf1/Cip1) plays a preventative role(s) in skin tumor formation. Surprisingly, moreover, we found that the level of p21(Waf1/Cip1) expression strikingly increased in the hair bulb and oscillated with a 3-week period correlating with hair follicle cycle progression. Notably, this was accompanied by the expression of p63 but not p53. This approach, together with the analysis of p21(Waf1/Cip1) knockout mice, has uncovered a novel role for the p21(Waf1/Cip1) gene in hair development. These data illustrate the unique utility of bioluminescence imaging in advancing our understanding of the timing and, hence, likely roles of specific gene expression in higher eukaryotes.
Toshiyuki Obata, Ichiro Yokota, Kazuhiro Yokoyama, Eiji Okamoto, Yoshiko Kanezaki, Yoshinori Tanaka, Hiroshi Maegawa, Kiyoshi Teshigawara, Fumiko Hirota, Tomoyuki Yuasa, Kazuhiro Kishi, Atsushi Hattori, Seiichi Hashida, Kazuhiko Masuda, Mitsuru Matsumoto, Toshio Matsumoto, Atsunori Kashiwagi and Yousuke Ebina : Soluble insulin receptor ectodomain is elevated in the plasma of patients with diabetes mellitus., Diabetes, Vol.56, No.8, 2028-2035, 2007.
(Summary)
Insulin binds to the alpha-subunit of the insulin receptor (IRalpha) and subsequently exerts its effects in the cells. The soluble ectodomains of several receptors have been found to circulate in the plasma. Therefore, we hypothesized that soluble human insulin receptor (hIR) ectodomain (alpha-subunit and a part of beta-subunit) may exist in the plasma of diabetic patients. We identified soluble hIR ectodomain in human plasma by a two-step purification followed by immunoblotting and gel-filtration chromatography. Furthermore, we established an hIRalpha-specific enzyme-linked immunosorbent assay and measured the plasma IRalpha levels in patients with diabetes. We also investigated this phenomenon in streptozotocin-induced diabetic hIR transgenic mice. Soluble hIRalpha, but not intact hIRbeta or whole hIR, exists in human plasma. The plasma IRalpha levels were significantly higher in type 1 (2.00 +/- 0.60 ng/ml; n = 53) and type 2 (2.26 +/- 0.80; n = 473) diabetic patients than in control subjects (1.59 +/- 0.40 ng/ml; n = 123 (P < 0.001 vs. control). Plasma IRalpha level was positively correlated with blood glucose level, and 10-20% of the insulin in plasma bound to hIRalpha. In the in vivo experiments using diabetic hIR transgenic mice, hyperglycemia was confirmed to increase the plasma hIRalpha level and the half-life estimated to be approximately 6 h. We propose that the increased soluble IR ectodomain level appears to be a more rapid glycemic marker than A1C or glycoalbumin.
Yoko Hamazaki, Harumi Fujita, Takashi Kobayashi, Yongwon Choi, Hamish S Scott, Mitsuru Matsumoto and Nagahiro Minato : Medullary thymic epithelial cells expressing Aire represent a unique lineage derived from cells expressing claudin, Nature Immunology, Vol.8, No.3, 304-311, 2007.
(Summary)
The autoimmune regulator Aire is expressed in a small proportion of medullary thymic epithelial cells (mTECs) and is crucial in the induction of central T cell tolerance. The origin and development of Aire(+) mTECs, however, are not well understood. Here we demonstrate that the tight-junction components claudin-3 and claudin-4 (Cld3,4) were 'preferentially' expressed in Aire(+) mTECs. In early ontogeny, Cld3,4(hi) TECs derived from the most apical layer of the stratified thymic anlage first expressed known mTEC markers such as UEA-1 ligand and MTS10. We provide evidence that such Cld3,4(hi) UEA-1(+) TECs represented the initial progenitors specified for Aire(+) mTECs, whose development crucially required NF-kappaB-inducing kinase and the adaptor molecule TRAF6. Our results suggest that Aire(+) mTECs represent terminally differentiated cells in a unique lineage arising during thymic organogenesis.
Shino Niki, Kiyotaka Oshikawa, Yasuhiro Mouri, Fumiko Hirota, Akemi Matsushima, Masashi Yano, Hongwei Han, Yoshimi Bando, Keisuke Izumi, Masaki Matsumoto, Keiichi I. Nakayama, Noriyuki Kuroda and Mitsuru Matsumoto : Alteration of intra-pancreatic target-organ specificity by abrogation of Aire in NOD mice, The Journal of Clinical Investigation, Vol.116, No.5, 1292-1301, 2006.
(Summary)
Factors that determine the spectrum of target organs involved in autoimmune destruction are poorly understood. Although loss of function of autoimmune regulator (AIRE) in thymic epithelial cells is responsible for autoimmunity, the pathogenic roles of AIRE in regulating target-organ specificity remain elusive. In order to gain insight into this issue, we have established NOD mice, an animal model of type 1 diabetes caused by autoimmune attack against beta cell islets, in which Aire has been abrogated. Remarkably, acinar cells rather than beta cell islets were the major targets of autoimmune destruction in Aire-deficient NOD mice, and this alteration of intra-pancreatic target-organ specificity was associated with production of autoantibody against pancreas-specific protein disulfide isomerase (PDIp), an antigen expressed predominantly by acinar cells. Consistent with this pathological change, the animals were resistant to the development of diabetes. The results suggest that Aire not only is critical for the control of self-tolerance but is also a strong modifier of target-organ specificity through regulation of T cell repertoire diversification. We also demonstrated that transcriptional expression of PDIp was retained in the Aire-deficient NOD thymus, further supporting the concept that Aire may regulate the survival of autoreactive T cells beyond transcriptional control of self-protein expression in the thymus.
Dan Kinoshita, Fumiko Hirota, Tsuneyasu Kaisho, Michiyuki Kasai, Keisuke Izumi, Yoshimi Bando, Yasuhiro Mouri, Akemi Matsushima, Shino Niki, Hongwei Han, Kiyotaka Oshikawa, Noriyuki Kuroda, Masahiko Maegawa, Minoru Irahara, Kiyoshi Takeda, Shizuo Akira and Mitsuru Matsumoto : Essential role of IkappaB kinase alpha in thymic organogenesis required for the establishment of self-tolerance., The Journal of Immunology, Vol.176, No.7, 3995-4002, 2006.
(Summary)
IkappaB kinase (IKK) alpha exhibits diverse biological activities through protein kinase-dependent and -independent functions, the former mediated predominantly through a noncanonical NF-kappaB activation pathway. The in vivo function of IKKalpha, however, still remains elusive. Because a natural strain of mice with mutant NF-kappaB-inducing kinase (NIK) manifests autoimmunity as a result of disorganized thymic structure with abnormal expression of Rel proteins in the thymic stroma, we speculated that the NIK-IKKalpha axis might constitute an essential step in the thymic organogenesis that is required for the establishment of self-tolerance. An autoimmune disease phenotype was induced in athymic nude mice by grafting embryonic thymus from IKKalpha-deficient mice. The thymic microenvironment that caused autoimmunity in an IKKalpha-dependent manner was associated with defective processing of NF-kappaB2, resulting in the impaired development of thymic epithelial cells. Thus, our results demonstrate a novel function for IKKalpha in thymic organogenesis for the establishment of central tolerance that depends on its protein kinase activity in cooperation with NIK.
Katsuaki Hoshino, Takahiro Sugiyama, Mitsuru Matsumoto, Takashi Tanaka, Masuyoshi Saito, Hiroaki Hemmi, Osamu Ohara, Shizuo Akira and Tsuneyasu Kaisho : IκB kinase-α is critical for interferon-α production induced by Toll-like receptors 7 and 9, Nature, Vol.440, No.7086, 949-953, 2006.
(Summary)
The Toll-like receptor (TLR) family has important roles in microbial recognition and dendritic cell activation. TLRs 7 and 9 can recognize nucleic acids and trigger signalling cascades that activate plasmacytoid dendritic cells to produce interferon-alpha (IFN-alpha) (refs 7, 8). TLR7/9-mediated dendritic cell activation is critical for antiviral immunity but also contributes to the pathogenesis of systemic lupus erythematosus, a disease in which serum IFN-alpha levels are elevated owing to plasmacytoid dendritic cell activation. TLR7/9-induced IFN-alpha induction depends on a molecular complex that contains a TLR adaptor, MyD88, and IFN regulatory factor 7 (IRF-7) (refs 10-14), but the underlying molecular mechanisms are as yet unknown. Here we show that IkappaB kinase-alpha (IKK-alpha) is critically involved in TLR7/9-induced IFN-alpha production. TLR7/9-induced IFN-alpha production was severely impaired in IKK-alpha-deficient plasmacytoid dendritic cells, whereas inflammatory cytokine induction was decreased but still occurred. Kinase-deficient IKK-alpha inhibited the ability of MyD88 to activate the Ifna promoter in synergy with IRF-7. Furthermore, IKK-alpha associated with and phosphorylated IRF-7. Our results identify a role for IKK-alpha in TLR7/9 signalling, and highlight IKK-alpha as a potential target for manipulating TLR-induced IFN-alpha production.
Yoshinori Nagai, Toshihiko Kobayashi, Yuji Motoi, Kohtaroh Ishiguro, Sachiko Akashi, Shin-ichiroh Saitoh, Yutaka Kusumoto, Tsuneyasu Kaisho, Shizuo Akira, Mitsuru Matsumoto, Kiyoshi Takatsu and Kensuke Miyake : The radioprotective 105/MD-1 complex links TLR2 and TLR4/MD-2 in antibody response to microbial membranes, The Journal of Immunology, Vol.174, No.11, 7043-7049, 2005.
(Summary)
Low-affinity IgG3 Abs to microbial membranes are important for primary immune defense against microbes, but little is known about the importance of TLRs in their production. IgG3 levels were extremely low in mice lacking radioprotective 105 (RP105), a B cell surface molecule structurally related to TLRs. RP105(-/-) B cells proliferated poorly in response to not only the TLR4 ligand LPS but also TLR2 ligand lipoproteins, both of which mediate the immunostimulatory activity of microbial membranes. RP105(-/-) mice were severely impaired in hapten-specific Ab production against LPS or lipoproteins. CD138 (syndecan-1)-positive plasma cells were detected after lipid A injection in wild-type spleen but much less in RP105(-/-) spleen. RP105 ligation in vivo induced plasma cell differentiation. RP105 expression was approximately 3-fold higher on marginal zone B cells than on follicular and B1 cells and was down-regulated on germinal center cells. These results demonstrate that a signal via RP105 is uniquely important for regulating TLR-dependent Ab production to microbial membranes.
Taishin Akiyam, Shiori Maeda, Sayaka Yamane, Kaori Ogino, Michiyuki Kasai, Fumiko Kajiura, Mitsuru Matsumoto and Jun-ichiro Inoue : Dependence of self-tolerance on TRAF6-directed development of thymic stroma, Science, Vol.308, No.5719, 248-251, 2005.
(Summary)
The microenvironments of the thymus are generated by thymic epithelial cells (TECs) and are essential for inducing immune self-tolerance or developing T cells. However, the molecular mechanisms that underlie the differentiation of TECs and thymic compartmentalization are not fully understood. Here we show that deficiency in the tumor necrosis factor receptor-associated factor (TRAF) 6 results in disorganized distribution of medullary TECs (mTECs) and the absence of mature mTECs. Engraftment of thymic stroma of TRAF6-/- embryos into athymic nude mice induced autoimmunity. Thus, TRAF6 directs the development of thymic stroma and represents a critical point of regulation for self-tolerance and autoimmunity.
Noriyuki Kuroda, Tasuku Mitani, Naoki Takeda, Naozumi Ishimaru, Rieko Arakaki, Yoshio Hayashi, Yoshimi Bando, Keisuke Izumi, Takeshi Takahashi, Takashi Nomura, Shimon Sakaguchi, Tomoo Ueno, Yousuke Takahama, Daisuke Uchida, Shijie Sun, Fumiko Kajiura, Yasuhiro Mouri, Hongwei Han, Akemi Matsushima, Gen Yamada and Mitsuru Matsumoto : Development of autoimmunity against transcriptionally unrepressed target antigen in the thymus of Aire-deficient mice, The Journal of Immunology, Vol.174, No.4, 1862-1870, 2005.
(Summary)
Autoimmune regulator (AIRE) gene mutation is responsible for the development of organ-specific autoimmune disease with monogenic autosomal recessive inheritance. Although Aire has been considered to regulate the elimination of autoreactive T cells through transcriptional control of tissue-specific Ags in thymic epithelial cells, other mechanisms of AIRE-dependent tolerance remain to be investigated. We have established Aire-deficient mice and examined the mechanisms underlying the breakdown of self-tolerance. The production and/or function of immunoregulatory T cells were retained in the Aire-deficient mice. The mice developed Sjogren's syndrome-like pathologic changes in the exocrine organs, and this was associated with autoimmunity against a ubiquitous protein, alpha-fodrin. Remarkably, transcriptional expression of alpha-fodrin was retained in the Aire-deficient thymus. These results suggest that Aire regulates the survival of autoreactive T cells beyond transcriptional control of self-protein expression in the thymus, at least against this ubiquitous protein. Rather, Aire may regulate the processing and/or presentation of self-proteins so that the maturing T cells can recognize the self-Ags in a form capable of efficiently triggering autoreactive T cells. With the use of inbred Aire-deficient mouse strains, we also demonstrate the presence of some additional factor(s) that determine the target-organ specificity of the autoimmune disease caused by Aire deficiency.
Tomoyuki Yuasa, Rei Kakuhata, Kazuhiro Kishi, Toshiyuki Obata, Yasuo Shinohara, Yoshimi Bando, Keisuke Izumi, Fumiko Kajiura, Mitsuru Matsumoto and Yousuke Ebina : Platelet-derived growth factor stimulates glucose transport in skeletal muscles of transgenic mice specifically expressing PDGF receptor in the muscle, but does not affect blood glucose levels, Diabetes, Vol.53, No.11, 2776-2786, 2004.
(Summary)
Insulin stimulates the disposal of blood glucose into skeletal muscle and adipose tissues by the translocation of GLUT4 from intracellular pools to the plasma membrane, and consequently the concentration of blood glucose levels decreases rapidly in vivo. Phosphatidylinositol (PI) 3-kinase and Akt play a pivotal role in the stimulation of glucose transport by insulin, but detailed mechanisms are unknown. We and others reported that not only insulin but also platelet-derived growth factor (PDGF) and epidermal growth factor facilitate glucose uptake through GLUT4 translocation by activation of PI 3-kinase and Akt in cultured cells. However, opposite results were also reported. We generated transgenic mice that specifically express the PDGF receptor in skeletal muscle. In these mice, PDGF stimulated glucose transport into skeletal muscle in vitro and in vivo. Thus, PDGF apparently shares with insulin some of the signaling molecules needed for the stimulation of glucose transport. The degree of glucose uptake in vivo reached approximately 60% of that by insulin injection in skeletal muscle, but blood glucose levels were not decreased by PDGF in these mice. Therefore, PDGF-induced disposal of blood glucose into skeletal muscle is insufficient for rapid decrease of blood glucose levels.
Hiroko Akiyoshi, Shigetsugu Hatakeyama, Jukka Pitkanen, Yasuhiro Mouri, Vassilis Doucas, Jun Kudoh, Kyoko Tsurugaya, Daisuke Uchida, Akemi Matsushima, Kiyotaka Oshikawa, Keiichi I. Nakayama, Nobuyoshi Shimizu, Part Peterson and Mitsuru Matsumoto : Subcellular expression of autoimmune regulator is organized in a spatiotemporal manner, The Journal of Biological Chemistry, Vol.279, No.32, 33984-33991, 2004.
Takashi Sakai, Li Liu, Xichuan Teng, Rika Mukai-Sakai, Hidenori Shimada, Ryuji Kaji, Tasuku Mitani, Mitsuru Matsumoto, Kazunori Toida, Kazunori Ishimura, Yuji Shishido, Tak W. Mak and Kiyoshi Fukui : Nucling Recruits Apaf-1/Pro-caspase-9 Complex for the Induction of Stress-Induced Apoptosis, The Journal of Biological Chemistry, Vol.279, No.39, 41131-41140, 2004.
(Summary)
Nucling is a novel protein isolated from murine embryonal carcinoma cells with an up-regulated expression during cardiac muscle differentiation. We show here that Nucling was up-regulated by proapoptotic stimuli and important for the induction of apoptosis after cytotoxic stress. We further demonstrated that overexpressed Nucling was able to induce apoptosis. In Nucling-deficient cells, the expression levels of Apaf-1 and cytochrome c, which are the major components of an apoptosis-promoting complex named apoptosome, were both down-regulated under cellular stress. A deficiency of Nucling also conferred resistance to apoptotic stress on the cell. After UV irradiation, Nucling was shown to reside in an Apaf-1/pro-caspase-9 complex, suggesting that Nucling might be a key molecule for the formation and maintenance of this complex. Nucling induced translocation of Apaf-1 to the nucleus, thereby distributing the Nucling/Apaf-1/pro-caspase-9 complex to the nuclear fraction. These findings suggest that Nucling recruits and transports the apoptosome complex during stress-induced apoptosis.
Yoshiko Kanezaki, Toshiyuki Obata, Rie Matsushima, Asako Minami, Tomoyuki Yuasa, Kazuhiro Kishi, Yoshimi Bando, Hisanori Uehara, Keisuke Izumi, Tasuku Mitani, Mitsuru Matsumoto, Yukari Takeshita, Yutaka Nakaya, Toshio Matsumoto and Yousuke Ebina : KATP Channel Knockout Mice Crossbred with Transgenic Mice Expressing a Dominant-Negative Form of Human Insulin Receptor have Glucose Intolerance but not Diabetes, Endocrine Journal, Vol.51, No.2, 133-144, 2004.
(Summary)
Impaired insulin secretion and insulin resistance are thought to be two major causes of type 2 diabetes mellitus. There are two kinds of diabetic model mice: one is a K(ATP) channel knockout (Kir6.2KO) mouse which is defective in glucose-induced insulin secretion, and the other is a transgenic mouse expressing the tyrosine kinase-deficient (dominant-negative form of) human insulin receptor (hIR(KM)TG), and which has insulin resistance in muscle and fat. However, all of these mice have no evidence of overt diabetes. To determine if the double mutant Kir6.2KO/hIR(KM)TG mice would have diabetes, we generated mutant mice by crossbreeding, which would show both impaired glucose-induced insulin secretion and insulin resistance in muscle and fat. We report here that: 1) blood glucose levels of randomly fed and 6 h fasted double mutant (Kir6.2KO/hIR(KM)TG) mice were comparable with those of wild type mice; 2) in intraperitoneal glucose tolerance test (ipGTT), Kir6.2KO/hIR(KM)TG mice had an impaired glucose tolerance; and 3) during ipGTT, insulin secretion was not induced in either Kir6.2KO/hIR(KM)TG or Kir6.2KO mice, while the hIR(KM)TG mice showed a more prolonged insulin secretion than did wild type mice; 4) hyperinsulinemic euglycemic clamp test revealed that Kir6.2KO, Kir6.2KO/hIR(KM)TG and hIR(KM)TG mice, showed decreased whole-body glucose disposal compared with wild type mice; 5) Kir6.2KO, but not Kir6.2KO/hIR(KM)TG mice had some obesity and hyperleptinemia compared with wild type mice. Thus, the defects in glucose-induced insulin secretion (Kir6.2KO) and an insulin resistance in muscle and fat (hIR(KM)TG) were not sufficient to lead to overt diabetes.
Fumiko Kajiura, Shijie Sun, Takashi Nomura, Keisuke Izumi, Tomoo Ueno, Yoshimi Bando, Noriyuki Kuroda, Hongwei Han, Yi Li, Akemi Matsushima, Yousuke Takahama, Shimon Sakaguchi, Tasuku Mitani and Mitsuru Matsumoto : NF-κB-inducing kinase establishes self-tolerance in a thymic stroma-dependent manner, The Journal of Immunology, Vol.172, No.4, 2067-2075, 2004.
(Summary)
Physical contact between thymocytes and the thymic stroma is essential for T cell maturation and shapes the T cell repertoire in the periphery. Stromal elements that control these processes still remain elusive. We used a mouse strain with mutant NF-kappaB-inducing kinase (NIK) to examine the mechanisms underlying the breakdown of self-tolerance. This NIK-mutant strain manifests autoimmunity and disorganized thymic structure with abnormal expression of Rel proteins in the stroma. Production of immunoregulatory T cells that control autoreactive T cells was impaired in NIK-mutant mice. The autoimmune disease seen in NIK-mutant mice was reproduced in athymic nude mice by grafting embryonic thymus from NIK-mutant mice, and this was rescued by supply of exogenous immunoregulatory T cells. Impaired production of immunoregulatory T cells by thymic stroma without normal NIK was associated with altered expression of peripheral tissue-restricted Ags, suggesting an essential role of NIK in the thymic microenvironment in the establishment of central tolerance.
Daisuke Uchida, Shigetsugu Hatakeyama, Akemi Matsushima, Hongwei Han, Satoshi Ishido, Hak Hotta, Jun Kudoh, Nobuyuki Shimizu, Vassilis Doucas, Keiichi I Nakayama, Noriyuki Kuroda and Mitsuru Matsumoto : AIRE functions as an E3 ubiquitin ligase, The Journal of Experimental Medicine, Vol.199, No.2, 166-172, 2004.
(Summary)
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.
A tyrosine kinase adaptor protein containing pleckstrin homology and SH2 domains (APS) is rapidly and strongly tyrosine phosphorylated by insulin receptor kinase upon insulin stimulation. The function of APS in insulin signaling has heretofore remained unknown. APS-deficient (APS(-/-)) mice were used to investigate its function in vivo. The blood glucose-lowering effect of insulin, as assessed by the intraperitoneal insulin tolerance test, was increased in APS(-/-) mice. Plasma insulin levels during fasting and in the intraperitoneal glucose tolerance test were lower in APS(-/-) mice. APS(-/-) mice showed an increase in the whole-body glucose infusion rate as assessed by the hyperinsulinemic-euglycemic clamp test. These findings indicated that APS(-/-) mice exhibited increased sensitivity to insulin. However, overexpression of wild-type or dominant-negative APS in 3T3L1 adipocytes did not affect insulin receptor numbers, phosphorylations of insulin receptor, insulin receptor substrate-1, or Akt and mitogen-activated protein kinase. The glucose uptake and GLUT4 translocation were not affected by insulin stimulation in these cells. Nevertheless, the insulin-stimulated glucose transport in isolated adipocytes of APS(-/-) mice was increased over that of APS(+/+) mice. APS(-/-) mice also showed increased serum levels of leptin and adiponectin, which might explain the increased insulin sensitivity of adipocytes.
Mitsuru Matsumoto, Takuji Yamada, Steven K. Yoshinaga, Tom Boone, Tom Horan, Shigeru Fujita, Yi Li and Tasuku Mitani : Essential role of NF-kappaB-inducing kinase in T cell activation through the TCR/CD3 pathway, The Journal of Immunology, Vol.169, No.3, 1151-1158, 2002.
(Summary)
NF-kappa B-inducing kinase (NIK) is involved in lymphoid organogenesis in mice through lymphotoxin-beta receptor signaling. To clarify the roles of NIK in T cell activation through TCR/CD3 and costimulation pathways, we have studied the function of T cells from aly mice, a strain with mutant NIK. NIK mutant T cells showed impaired proliferation and IL-2 production in response to anti-CD3 stimulation, and these effects were caused by impaired NF-kappa B activity in both mature and immature T cells; the impaired NF-kappa B activity in mature T cells was also associated with the failure of maintenance of activated NF-kappa B. In contrast, responses to costimulatory signals were largely retained in aly mice, suggesting that NIK is not uniquely coupled to the costimulatory pathways. When NIK mutant T cells were stimulated in the presence of a protein kinase C (PKC) inhibitor, proliferative responses were abrogated more severely than in control mice, suggesting that both NIK and PKC control T cell activation in a cooperative manner. We also demonstrated that NIK and PKC are involved in distinct NF-kappa B activation pathways downstream of TCR/CD3. These results suggest critical roles for NIK in setting the threshold for T cell activation, and partly account for the immunodeficiency in aly mice.
(Keyword)
Animals / I-kappa B Kinase / Interleukin-2 / Lymphocyte Activation / Mice / NF-kappa B / Protein Kinase C / Protein-Serine-Threonine Kinases / Proto-Oncogene Proteins / Receptor-CD3 Complex, Antigen, T-Cell / T-Lymphocytes / Tetradecanoylphorbol Acetate / Transcription Factors
(Link to Search Site for Scientific Articles)
● PubMed @ National Institutes of Health, US National Library of Medicine (PMID): 12133934
Akemi Matsushima, Tsuneyasu Kaisho, Paul D. Rennert, Hiroyasu Nakano, Kyoko Kurosawa, Daisuke Uchida, Kiyoshi Takeda, Shizuo Akira and Mitsuru Matsumoto : Essential role of NF-kappaB-inducing kinase and IkappaB-kinase alpha in NF-kappaB activation through lymphotoxin-beta receptor, but not through TNF receptor-I, The Journal of Experimental Medicine, Vol.193, No.5, 631-636, 2001.
(Summary)
Both nuclear factor (NF)-kappaB-inducing kinase (NIK) and inhibitor of kappaB (IkappaB) kinase (IKK) have been implicated as essential components for NF-kappaB activation in response to many external stimuli. However, the exact roles of NIK and IKKalpha in cytokine signaling still remain controversial. With the use of in vivo mouse models, rather than with enforced gene-expression systems, we have investigated the role of NIK and IKKalpha in signaling through the type I tumor necrosis factor (TNF) receptor (TNFR-I) and the lymphotoxin beta receptor (LTbetaR), a receptor essential for lymphoid organogenesis. TNF stimulation induced similar levels of phosphorylation and degradation of IkappaBalpha in embryonic fibroblasts from either wild-type or NIK-mutant mice. In contrast, LTbetaR stimulation induced NF-kappaB activation in wild-type mice, but the response was impaired in embryonic fibroblasts from NIK-mutant and IKKalpha-deficient mice. Consistent with the essential role of IKKalpha in LTbetaR signaling, we found that development of Peyer's patches was defective in IKKalpha-deficient mice. These results demonstrate that both NIK and IKKalpha are essential for the induction of NF-kappaB through LTbetaR, whereas the NIK-IKKalpha pathway is dispensable in TNFR-I signaling.
(Keyword)
Animals / Antigens, CD / Cells, Cultured / DNA-Binding Proteins / Fibroblasts / I-kappa B Kinase / I-kappa B Proteins / Lymphotoxin beta Receptor / Mice / Mice, Inbred C57BL / Mice, Mutant Strains / NF-kappa B / Peyer's Patches / Phosphorylation / Protein-Serine-Threonine Kinases / Receptors, Tumor Necrosis Factor / Receptors, Tumor Necrosis Factor, Type I / Signal Transduction / Transfection / Tumor Necrosis Factor-alpha
Masaya Uwai, Yasuhito Terui, Yuji Mishima, Hiroshi Tomizuka, Masayuki Ikeda, Takehito Itoh, Masaki Mori, Masuzu Ueda, Rie Inoue, Muneo Yamada, Hirotoshi Hayasawa, Takahiko Horiuchi, Yoshiyuki Niho, Mitsuru Matsumoto, Yukihito Ishizaka, Kazuma Ikeda, Keiya Ozawa and Kiyohiko Hatake : A new apoptotic pathway for the complement factor B-derived fragment Bb, Journal of Cellular Physiology, Vol.185, No.2, 280-292, 2000.
61.
Takuji Yamada, Tasuku Mitani, Kazuko YORITA, Daisuke Uchida, Akemi Matsushima, Kikue Iwamasa, Shigeru Fujita and Mitsuru Matsumoto : Abnormal Immune Function of Hemopoietic Cells from Alymphoplasia (aly) Mice, a Natural Strain with Mutant NF-kappaB-Inducing Kinase, The Journal of Immunology, Vol.165, No.2, 804-812, 2000.
(Summary)
Alymphoplasia (aly) mice, a natural strain with a mutant NF-kappa B-inducing kinase (NIK) gene, manifest a unique phenotype; they lack lymph nodes and Peyer's patches, have a disturbed spleen architecture, and exhibit defects in both Ab and cellular immune responses. Although a stromal defect caused by impaired lymphotoxin-beta receptor signaling accounts for their abnormal lymphoid organogenesis, the exact mechanisms underlying the development of immunodeficiency in aly mice are poorly understood. We therefore investigated the contribution of hemopoietic cells with the aly NIK mutation to the development of immunodeficiency. Transfer of aly/aly bone marrow cells into aly/+ mice resulted in poorly developed B cell follicles and lack of support for the development of germinal centers and isotype switching, indicating that the hemopoietic cells of aly mice contain an autonomous defect. However, follicular dendritic cell clusters were maintained in the spleens of these bone marrow chimeras, suggesting that the lack of follicular dendritic cell clusters in aly mice is probably due to the stromal defect. The aly mice lacked marginal zone B cells in their spleens, and aly/aly B cells showed an impaired proliferative response after in vitro stimulation. IL-2 production by activated T cells was also impaired. By contrast, the dendritic cells of aly mice exhibited grossly normal development and function. Supporting the concept of an autonomous cell defect, Rel protein expression was altered in aly/aly spleens. Thus, the aly NIK mutation affects hemopoietic cell function in an intrinsic fashion and, together with the stromal defect, may contribute to the development of immunodeficiency in aly mice.
Hiroyasu Nakano, Sachiko Sakon, Haruhiko Koseki, Toshitada Takemori, Kurisu Tada, Mitsuru Matsumoto, Eiko Munechika, Tsuyoshi Sasaki, Takuji Shirasawa, Hisaya Akiba, Tetsuji Kobata, Sybil M. Santee, Carl F. Ware, Paul D. Rennert, Masaru Taniguchi, Hideo Yagita and Ko Okumura : Targeted disruption of Traf5 gene causes defects in CD40- and CD27-mediated lymphocyte activation, Proceedings of the National Academy of Sciences of the United States of America, Vol.96, No.17, 9803-9808, 1999.
63.
Mitsuru Matsumoto, Kikue Iwamasa, Paul D. Rennert, Takuji Yamada, Rika Suzuki, Akemi Matsushima, Masaru Okabe, Shigeru Fujita and Minesuke Yokoyama : Involvement of distinct cellular compartments in the abnormal lymphoid organogenesis in lymphotoxin-alpha-deficient mice and alymphoplasia (aly) mice defined by the chimeric analysis, The Journal of Immunology, Vol.163, No.3, 1584-1591, 1999.
(Summary)
Both lymphotoxin-alpha (LTalpha)-deficient mice and alymphoplasia (aly) mice, a natural mutant strain, manifest a quite similar phenotype: lack of lymph nodes (LN) and Peyer's patches (PP), with disturbed spleen architecture. The mechanisms underlying the defective lymphoid organogenesis in these mice were investigated by generating aggregation chimeras; ex vivo fused morulae were implanted into pseudo-pregnant host females and allowed to develop to term. Chimeric mice between LTalpha-deficient mice and wild-type mice restored LN and PP almost completely, suggesting that LTalpha expressed by circulating bone marrow-derived cells is essential for lymphoid organogenesis as well as for organization of spleen architecture. By contrast, chimeric mice between aly mice and wild-type mice showed only limited restoration of LN and PP. This suggests that the putative aly gene product does not act as a circulating ligand for lymphoid organogenesis, like LTalpha. Rather, abnormal development of lymphoid organs in aly mice seems most likely due to the defective development of the incipient stromal cells of the LN and PP. Supporting this hypothesis, up-regulation of VCAM-1 on aly mouse embryonic fibroblasts by signals through LTbetaR, which is exclusively expressed by nonlymphoid cells, was disturbed. These studies demonstrate that LTalpha and the putative aly gene product together control lymphoid organogenesis with a close mechanistic relationship in their biochemical pathways through governing the distinct cellular compartments, the former acting as a circulating ligand and the latter as a LTbetaR-signaling molecule expressed by the stroma of the lymphoid organs.
Yang-Xin Fu, Guangming Huang, Mitsuru Matsumoto, Hector Molina and David D Chaplin : Independent signals regulate development of primary and secondary follicle structure in spleen and mesenteric lymph node, Proceedings of the National Academy of Sciences of the United States of America, Vol.94, No.11, 5739-5743, 1997.
65.
Yang-Xin Fu, Hector Molina, Mitsuru Matsumoto, Guangming Huang, Jingjuan Min and David D Chaplin : Lymphotoxin-alpha (LTalpha) supports development of splenic follicular structure that is required for IgG responses, The Journal of Experimental Medicine, Vol.185, No.12, 2111-2120, 1997.
66.
Mitsuru Matsumoto, Yang-Xin Fu, Hector Molina, Guangming Huang, Jinho Kim, Dori A. Thomas, Moon H. Nahm and David D. Chaplin : Distinct roles of lymphotoxin-alpha and the type I TNF receptor in the establishment of follicular dendritic cells from non-bone marrow-derived cells, The Journal of Experimental Medicine, Vol.186, No.12, 1997-2004, 1997.
67.
Mitsuru Matsumoto, Wataru Fukuda, Antonella Circolo, Joseph Goellner, Jena Strauss-Schoenberger, Xuefeng Wang, Shigeru Fujita, Tunde Hidvegi, David D Chaplin and Harvey R Colten : Abrogation of the alternative complement pathway by targeted deletion of murine factor B, Proceedings of the National Academy of Sciences of the United States of America, Vol.94, 8720-8725, 1997.
68.
Mitsuru Matsumoto, Stanley F Lo, Cynthia JL Carruthers, Jingjuan Min, Sanjeev Mariathasan, Guangming Huang, David R Plas, Steven M Martin, Raif S Geha, Moon H Nahm and David D Chaplin : Affinity maturation without germinal centres in lymphotoxin-alpha-deficient mice, Nature, Vol.382, No.6590, 462-466, 1996.
(Summary)
Affinity maturation by somatic hypermutation is thought to occur within germinal centres. Mice deficient in lymphotoxin-alpha (LT alpha-/- mice) have no lymph nodes or Peyer's patches, and fail to form germinal centres in the spleen. We tested whether germinal centres are essential for maturation of antibody responses to T-cell-dependent antigens. LT alpha-/- mice immunized with low doses of (4-hydroxy-3-nitrophenyl)acetyl-ovalbumin (NP-OVA) showed dramatically impaired production of high-affinity anti-NP IgG1. However, LT alpha-/- mice immunized with high doses of NP-OVA, even though they failed to produce germinal centres, manifested a high-affinity anti-NP IgG1 response similar to wild-type mice. Furthermore, when LT alpha-/- mice were multiply immunized with high doses of NP-OVA, the predominantly expressed anti-NP VH gene segment VH186.2 showed somatic mutations typical of affinity maturation. Thus, B-cell memory and affinity maturation are not absolutely dependent on the presence of germinal centres.
Mitsuru Matsumoto, Sanjeev Mariathasan, Moon H Nahm, Ferenc Baranyay, Jacques J Peschon and David D Chaplin : Role of lymphotoxin and the type I TNF receptor in the formation of germinal centers, Science, Vol.271, No.5253, 1289-1291, 1996.
(Summary)
In mice deficient in either lymphotoxin-alpha (LT-alpha) or the type I tumor necrosis factor (TNF) receptor, but not the type II TNF receptor, germinal centers failed to develop in peripheral lymphoid organs. Germinal center formation was restored in LT-alpha-deficient mice by transplantation of normal bone marrow, indicating that the LT-alpha-expressing cells required to establish this lymphoid structure are derived from bone marrow.
(Keyword)
Animals / Bone Marrow Cells / Bone Marrow Transplantation / Gene Targeting / Germinal Center / Immunization / Lymphotoxin-alpha / Mice / Receptors, Tumor Necrosis Factor / Spleen
Mitsuru Matsumoto, Yoshiki Tange, Takanori Okada, Yasuhiro Inoue, Takahiko Horiuchi, Yuzuru Kobayashi and Shigeru Fujita : Deletion in the 190 kDa antigen gene repeat region of Rickettsia rickettsii, Microbial Pathogenesis, Vol.20, No.1, 57-62, 1996.
(Summary)
The 190 kDa outer membrane protein of Rickettsia rickettsii is a major immunodominant protective antigen which contains 13 tandem nearly identical repeating sequences. We have identified a deletion in the 190 kDa antigen gene of R. rickettsii strain Smith maintained in the laboratory. The deletion occurred within the repeat region of this gene and the mutated repeat region corresponded to one repeating unit in size.
Sanjeev Mariathasan, Mitsuru Matsumoto, Ferenc Baranyay, Moon M Nahm, Osami Kanagawa and David D Chaplin : Absence of lymph nodes in lymphotoxin-alpha (LTalpha)-deficient mice is due to abnormal organ development, not defective lymphocyte migration, Journal of Inflammation, Vol.45, 72-78, 1995.
Minoru Matsumoto, Koichi Tsuneyama and Mitsuru Matsumoto : AIRE遺伝子と多腺性自己免疫症候群, The Medical Frontline, Vol.73, No.5, 688-692, May 2018.
5.
Mitsuru Matsumoto : Switching on the Aire conditioner, European Journal of Immunology, Vol.45, No.12, 3237-3240, Dec. 2015.
(Summary)
Aire has been cloned as the gene responsible for a hereditary type of organ-specific autoimmune disease. Aire controls the expression of a wide array of tissue-restricted Ags by medullary thymic epithelial cells (mTECs), thereby leading to clonal deletion and Treg-cell production, and ultimately to the establishment of self-tolerance. However, relatively little is known about the mechanism responsible for the control of Aire expression itself. In this issue of the European Journal of Immunology, Haljasorg et al. [Eur. J. Immunol. 2015. 45: 3246-3256] have reported the presence of an enhancer element for Aire that binds with NF-κB components downstream of the TNF receptor family member, RANK (receptor activator of NF-κB). The results suggest that RANK has a dual mode of action in Aire expression: one involving the promotion of mTEC differentiation and the other involving activation of the molecular switch for Aire within mature mTECs.
Mitsuru Matsumoto, Yumiko Nishikawa, Hitoshi Nishijima, Junko Morimoto, Minoru Matsumoto and Yasuhiro Mouri : Which model better fits the role of Aire in the establishment of self-tolerance: the transcription model or the maturation model?, Frontiers in Immunology, Vol.4, 1-4, Jul. 2013.
(Summary)
The discovery of Aire-dependent transcriptional control of many tissue-restricted self-antigen (TRA) genes in thymic epithelial cells in the medulla (medullary thymic epithelial cells, mTECs) has raised the intriguing question of how the single Aire gene can influence the transcription of such a large number of TRA genes within mTECs. From a mechanistic viewpoint, there are two possible models to explain the function of Aire in this action. In the first model, TRAs are considered to be the direct target genes of Aire's transcriptional activity. In this scenario, the lack of Aire protein within cells would result in the defective TRA gene expression, while the maturation program of mTECs would be unaffected in principle. The second model hypothesizes that Aire is necessary for the maturation program of mTECs. In this case, we assume that the mTEC compartment does not mature normally in the absence of Aire. If acquisition of the properties of TRA gene expression depends on the maturation status of mTECs, a defect of such an Aire-dependent maturation program in Aire-deficient mTECs can also result in impaired TRA gene expression. In this brief review, we will focus on these two contrasting models for the roles of Aire in controlling the expression of TRAs within mTECs.
Mitsuru Matsumoto : Contrasting models for the roles of Aire in the differentiation program of epithelial cells in the thymic medulla, European Journal of Immunology, Vol.41, No.1, 12-17, Jan. 2011.
(Summary)
The current prevailing view regarding the role of Aire in self-tolerance is that it is involved in the transcriptional control of many tissue-restricted self-antigen genes in thymic epithelial cells in the medulla (mTECs); however, accumulating evidence also suggests that Aire has other roles, e.g. in mTEC differentiation, and furthermore that Aire can either promote or inhibit the mTEC differentiation program, i.e. Aire does not play a neutral role in mTEC differentiation. This review discusses when and how Aire plays an important role in controlling the organization of mTECs required for the expression of self-antigen genes.
Mitsuru Matsumoto : The role of autoimmune regulator (Aire) in the development of the immune system, Microbes and Infection, Vol.11, No.12, 928-934, Jul. 2009.
(Summary)
AIRE is the gene responsible for a rather rare hereditary type of autoimmune disease. The mechanism underlying the autoimmune pathogenesis caused by AIRE deficiency is a focus of intense research because it could provide clues to the fundamental question of how the immune system discriminates between self and non-self within the thymic microenvironment.
Mitsuru Matsumoto : Autoimmune regulator functions in autoimmunity control., Expert Review of Clinical Immunology, Vol.3, No.6, 891-900, Nov. 2007.
(Summary)
Mutation of the autoimmune regulator (AIRE) gene is responsible for the development of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED), an organ-specific autoimmune disease with monogenic autosomal recessive inheritance. AIRE is strongly expressed in thymic epithelial cells (TECs), and its structural features suggest that it probably acts as a transcriptional regulator. The role of Aire in the elimination of autoreactive T cells (i.e., negative selection) has been well demonstrated with the use of transgenic mouse models, although how Aire regulates this process in TECs has yet to be determined. Aire also regulates other features of autoimmunity, such as target-organ specificity of autoimmune destruction, suggesting that an understanding of the relationship between AIRE gene malfunction and the breakdown of self-tolerance promises to help unravel the pathogenesis of not only APECED but also other types of autoimmune diseases. Owing to this unique tolerogenic function of Aire, the cellular origin and developmental process of Aire-expressing TECs is now becoming an interesting field for intense research.
Mitsuru Matsumoto : Transcriptional regulation in thymic epithelial cells for the establishment of self tolerance, Archivum Immunologiae et Therapiae Experimentalis, Vol.55, No.1, 27-34, 2007.
(Summary)
Thymic epithelial cells (TECs) play pivotal roles in the establishment of self tolerance through critical dialogue with developing thymocytes. Unique actions of two transcriptional regulators within TECs, NF-kappaB-inducing kinase (NIK) and an autoimmune regulator (AIRE), for the establishment of self tolerance have recently been highlighted by studies using a strain of mouse bearing a natural mutation of the NIK gene (aly mice) and gene-targeted mice, respectively. Previous studies have demonstrated essential roles of NIK downstream of the lymphotoxin-beta receptor (LTbetaR), which is essential for the development of secondary lymphoid organs; aly mice lack all lymph nodes and Peyer's patches because of the defective LTbetaR signaling. Additional roles of NIK in thymic organogenesis downstream of LTPR, mainly through the developmental regulation of TECs, have now emerged, although the corresponding ligand(s) for LTbetaR participating in this action have not been fully characterized. In contrast, AIRE, a gene responsible for the development of an organ-specific autoimmune disease that demonstrates monogenic autosomal recessive inheritance, contributes to the establishment of self tolerance probably by controlling the expression of self antigens through yet undetermined molecular mechanisms. Thus, it is highly likely that a group of genes control self tolerance within TECs through unique and coordinated actions, and that an understanding of this process would help to unravel the pathogenesis of autoimmune disease.
Mitsuru Matsumoto : Role of TNF ligand and receptor family in the lymphoid organogenesis defined by gene targeting, The Journal of Medical Investigation : JMI, Vol.46, No.3,4, 141-150, Aug. 1999.
(Summary)
The molecular basis of lymphoid organogenesis has recently been elucidated using gene-targeted mice. Mice deficient in lymphotoxin-alpha (LT alpha) lack lymph nodes and Peyer's patches. The action of LT alpha in lymphoid organogenesis is mediated mostly by the membrane form of LT by a mechanism independent of TNF receptor I (TNFR-I) or II (TNFR-II). Additionally, follicular dendritic cell (FDC) clusters or germinal centers fail to develop in the spleen of LT alpha-deficient mice. Mice deficient in either TNFR-I or LT beta R also fail to develop splenic FDC clusters and germinal centers, indicating that signaling through both TNFR-I and LT beta R is required for the development of these structures. The mechanisms underlying the defective lymphoid organogenesis in LT alpha-deficient mice, together with a natural mutant strain, alymphoplasia (aly) mice, which manifest a quite similar phenotype to LT alpha-deficient mice, were investigated by generating aggregation chimeras. These studies demonstrate that LT alpha and the aly gene product together control lymphoid organogenesis with a close mechanistic relationship in their biochemical pathways through governing distinct cellular compartments; the former acting as a circulating ligand and the latter as a LT beta R-signaling molecule expressed by the stroma of the lymphoid organs.
(Keyword)
Animals / Antigens, CD / Gene Targeting / Ligands / Lymphoid Tissue / Lymphotoxin-alpha / Mice / Mice, Mutant Strains / Receptors, Tumor Necrosis Factor / Receptors, Tumor Necrosis Factor, Type I / Receptors, Tumor Necrosis Factor, Type II / Tumor Necrosis Factor-alpha
(Tokushima University Institutional Repository: 21957, PubMed: 10687308)
15.
Mitsuru Matsumoto, Yang-Xin Fu, Hector Molina and David D. Chaplin : Lymphotoxin-alpha deficient and TNF receptor-I-deficient mice define developmental and functional characteristics of germinal centers, Immunological Reviews, Vol.156, 137-144, 1997.
Proceeding of International Conference:
1.
Minoru Matsumoto, Mitsuru Matsumoto, Junko Morimoto and Hitoshi Nishijima : Tissue-specific Autoimmunity Controlled by Aire, a Gene Responsible for APECED, The 3rd International Congress on Rare Diseases, Berlin, Germany, Mar. 2020.
2.
Minoru Matsumoto, Hitoshi Nishijima, Ryuichiro Miyazawa, Junko Morimoto, Koichi Tsuneyama and Mitsuru Matsumoto : Characterization of Aire-expressing DCs with high-sensitivity and high-fidelity Aire-reporter strain, The 48th Annual Meeting of the Japanese Society for Immunology, Hamamatsu, Japan, Dec. 2019.
3.
Mitsuru Matsumoto, Junko Morimoto, Minoru Matsumoto, Koichi Tsuneyama, Ryuichiro Miyazawa and Hitoshi Nishijima : Aire-dependent establishment of self-tolerance, The 17th International Congress of Immunology, Beijing, Oct. 2019.
4.
Minoru Matsumoto and Mitsuru Matsumoto : Tissue-specific autoimmunity controlled by Aire in thymic and peripheral tolerance mechanisms, The 18th Awaji International Forum on Infection and Immunity, Awaji, Japan, Sep. 2019.
5.
Mitsuru Matsumoto, Junko Morimoto, Minoru Matsumoto, Koichi Tsuneyama, Ryuichiro Miyazawa and Hitoshi Nishijima : Aire-dependent establishment of self-tolerance studied by genetically modified mice, EMBO Workshop ThymE, Israel, May 2019.
6.
Minoru Matsumoto, Junko Morimoto, Mitsuru Matsumoto and Hitoshi Nishijima : Tissue-specific autoimmunity modified by Aire in thymic and peripheral tolerance, 5th International Congress on Controversies in Rheumatology and Autoimmunity (CORA), Italy, Mar. 2019.
7.
Mitsuru Matsumoto, Minoru Matsumoto, Junko Morimoto, Koichi Tsuneyama and Hitoshi Nishijima : Tissue-specific autoimmunity manipulated by Aire in thymic and peripheral tolerance, Immunology of Diabetes Society Congress 2018, London, Oct. 2018.
8.
Mitsuru Matsumoto, Junko Morimoto, Minoru Matsumoto and Hitoshi Nishijima : Aire controls in trans the production of medullary thymic epithelial cells expressing Ly6C/Ly6G, 5th European Congress of Immunology 2018, Amsterdam, Sep. 2018.
9.
Minoru Matsumoto, Hitoshi Nishijima, Junko Morimoto and Mitsuru Matsumoto : Distinct tissue-specific immune response revealed by manipulation of thymic and peripheral tolerance by Aire, 1st International Symposium for "Neo-self", Awaji, Japan, Jul. 2018.
10.
Mitsuru Matsumoto : Immune tolerance mediated by Aire, 1st International Symposium for "Neo-self", Awaji, Japan, Jul. 2018.
11.
Minoru Matsumoto, Hitoshi Nishijima, Junko Morimoto, Koichi Tsuneyama and Mitsuru Matsumoto : Manipulation of thymic and peripheral tolerance by AIRE defines distinct tissue-specific autoimmunity, 11th International Congress on Autoimmunity, Lisbon, Portugal, May 2018.
12.
Junko Morimoto, Yumiko Nishikawa, Naoki Kihara, Kazuyoshi Hosomichi, Hitoshi Nishijima and Mitsuru Matsumoto : Expression of Ly6C/6G defines a novel subset of medullary thymic epithelial cells, Immunology 2018 - AAI Annual Meeting, Austin, Texas, May 2018.
13.
Mitsuru Matsumoto, Junko Morimoto, Minoru Matsumoto, Koichi Tsuneyama and Hitoshi Nishijima : A novel Aire-dependent subset of mTECs with tolerogenic functions is defined by Ly6 family protein expression, ThymOz8, Heron Island, Australia, Mar. 2018.
14.
Minoru Matsumoto, Hitoshi Nishijima, Junko Morimoto, Koichi Tsuneyama and Mitsuru Matsumoto : Acquisition of the resistance to autoimmune diabetes by the expression of human AIRE in BM-derived APCs in NOD, ThymOz8, Heron Island, Australia, Mar. 2018.
15.
Junko Morimoto, Yumiko Nishikawa, Naoki Kihara, Kazuyoshi Hosomichi, Hitoshi Nishijima and Mitsuru Matsumoto : Expression of Ly6C/6G defines a novel subset of medullary thymic epithelial cells, 5th Annual Meeting of the International Cytokine and Interferon Society, Kanazawa, Japan, Oct. 2017.
16.
Junko Morimoto, Yumiko Nishikawa, Hitoshi Nishijima and Mitsuru Matsumoto : Expression of Ly6C/6G defines a novel Aire-dependent subset of medullary thymic epithelial cells with tolerogenic function, 12th International Congress on SLE, Melbourne, Australia, Mar. 2017.
17.
Hitoshi Nishijima, Yasuhiro Mouri, Junko Morimoto and Mitsuru Matsumoto : Paradoxical development of polymyositis-like autoimmunity through augmented expression of AIRE, 7th International Workshop of Kyoto T Cell Conference, Kyoto, Japan, Mar. 2017.
18.
Hitoshi Nishijima, Yasuhiro Mouri, Junko Morimoto and Mitsuru Matsumoto : Paradoxical development of polymyositis-like autoimmunity through augmented expression of human AIRE in mice, Controversies in Rheumatology and Autoimmunity 2017, Bologna, Italy, Mar. 2017.
19.
Hitoshi Nishijima and Mitsuru Matsumoto : Massive infiltration of macrophages and eosinophils in the muscle of mice overexpressing Aire in thymic and peripheral antigen-presenting cells, Cell Symposia - 100 Years of Phagocytosis, Sicily, Italy, Sep. 2016.
20.
Hitoshi Nishijima and Mitsuru Matsumoto : Augmented expression of the autoimmune regulator (AIRE) induces paradoxical development of polymyositis-like autoimmunity, Immunology 2016 - AAI Annual Meeting, Seattle, USA, May 2016.
21.
Hitoshi Nishijima and Mitsuru Matsumoto : Augmented expression of the autoimmune regulator (AIRE) induces paradoxical development of polymyositis-like autoimmunity in mice, The Fifth Bizan Immunology Symposium at University of Tokushima, Mar. 2016.
22.
Hiroshi Kawano, Hitoshi Nishijima, Junko Morimoto, Yasuhiro Mouri and Mitsuru Matsumoto : Aire expression is inherent to all medullary thymic epithelial cells during their differentiation program, The 4th European Congress of Immunology, Vienna, Austria, Sep. 2015.
23.
Mitsuru Matsumoto, Hiroshi Kawano, Hitoshi Nishijima, Junko Morimoto, Yasuhiro Mouri and Yasuhiko Nishioka : Aire expression is inherent properties of all medullary thymic epithelial cells during their differentiation program, Venice Thymus Meeting 2015, Venice, Italy, Apr. 2015.
24.
Hiroshi Kawano and Mitsuru Matsumoto : Unexpected disturbance of the development of medullary thymic epithelial cells at immature stages by the long-term ablation of Aire-expressing cells, Controversies in Rheumatology and Autoimmunity 2015, Sorrento, Italy, Mar. 2015.
25.
Mitsuru Matsumoto and Hitoshi Nishijima : Ectopic Aire expression in thymic cortex reveals inherent properties of Aire as a medullary stromal factor, European Society for Immunodeficiencies 2014, Prague, Czech Republic, Oct. 2014.
26.
Hitoshi Nishijima and Mitsuru Matsumoto : Ectopic Aire expression in thymic cortex reveals inherent properties of Aire as a stromal factor for medullary thymic microenvironment, 9th International Symposium of the Institute Network, Jun. 2014.
27.
Hitoshi Nishijima, Junko Morimoto, Hiroshi Kawano, Yasuhiro Mouri, Koichi Ikuta and Mitsuru Matsumoto : Ectopic Aire expression in thymic cortex reveals inherent properties of Aire as a stromal factor for medullary thymic microenvironment, ThymOz 7, Heron Island, Australia, Apr. 2014.
28.
Mitsuru Matsumoto, Junko Morimoto, Hiroshi Kawano, Yasuhiro Mouri and Hitoshi Nishijima : Ectopic Aire expression in thymic cortex reveals inherent properties of Aire as a stromal factor for medullary microenvironment, 9th International Congress on Autoimmunity, Nice, France, Mar. 2014.
29.
Hitoshi Nishijima, Junko Morimoto, Yasuhiro Mouri, Yumiko Nishikawa, Kouichi Ikuta and Mitsuru Matsumoto : Ectopic Aire expression in thymic cortex reveals inherent properties of Aire as a stromal factor within medullary microenvironment, 第3回 眉山免疫シンポジウム, Tokushima, Feb. 2014.
30.
Mitsuru Matsumoto : Aire-dependent organization of thymic microenvironment for the establishment of self-tolerance, WIP-Workshop, Munich, Germany, Oct. 2013.
31.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri and Hitoshi Nishijima : Temporal lineage tracing of Aire-expressing cells reveals a requirement of Aire for their maturation program, 15th ICI2013, Milan, Italy, Aug. 2013.
32.
Mitsuru Matsumoto, Yumiko Nishikawa, Hitoshi Nishijima, Minoru Matsumoto, Fumiko Hirota, Satoru Takahashi, Hervé Luche, Hans Fehling Joerg and Yasuhiro Mouri : Temporal lineage tracing of Aire-expressing cells reveals a requirement for Aire in their maturation program, The 6th International Workshop of Kyoto T Cell Conference, Kyoto, Japan, Jun. 2013.
33.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri and Hitoshi Nishijima : Temporal fate-mapping study reveals essential roles of Aire in the maturation program of Aire-expressing cell lineage, Immunology 2013 - AAI Annual Meeting, Honolulu, Hawaii, May 2013.
34.
Yumiko Nishikawa, Yasuhiro Mouri, Hitoshi Nishijima and Mitsuru Matsumoto : Temporal lineage tracing of Aire-expressing cells reveals a requirement of Aire for their full maturation program, Controversies in Rheumatology and Autoimmunity (CORA 2011), Budapest, Hungary, Apr. 2013.
35.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri and Hitoshi Nishijima : Essential roles of Aire in the maturation program of Aire-expressing cell lineage(s) revealed by temporal fate-mapping approach, The 10th International Conference on New Trends in Immunosuppression and Immunotherapy, Barcelona, Spain, Mar. 2013.
36.
Mitsuru Matsumoto : Aire-dependent organization of thymic microenvironment for the establishment of self-tolerance, 2013 SKKU International Symposium on Molecular Medicine: New Trends in Cancer and Autoimmunity Research, Suwon, Republic of Korea, Feb. 2013.
37.
Mitsuru Matsumoto : Control of chronic inflammation through elucidation of organ-specific autoimmune disease mechanisms, JST-CREST International Symposium, Tokyo, Japan, Feb. 2013.
38.
Mitsuru Matsumoto : Aire-dependent organization of thymic microenvironment for the establishment of self tolerance, Centennial of Hashimoto Disease, Fukuoka, Japan, Dec. 2012.
39.
Yumiko Nishikawa and Mitsuru Matsumoto : Temporal fate-mapping approach reveals a requirement of Aire for the full maturation program of thymic epithelial cells in the medulla, International Symposium on Genetic and Epigenetic Control of Cell Fate, Kyoto, Japan, Nov. 2012.
40.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri, Fumiko Hirota and Hitoshi Nishijima : Temporal fate-mapping reveals essential roles of Aire in the maturation program of mTECs, ThymUS 2012, Florida, USA, Nov. 2012.
41.
Mitsuru Matsumoto : Aire-dependent organization of thymic microenvironment for the establishment of self-tolerance, IEIIS/HIS 2012, Tokyo, Japan, Oct. 2012.
42.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri and Hitoshi Nishijima : Functional analysis of Aire, a gene responsible for the hereditary type of autoimmune disease, Days of Molecular Medicine 2012, Wien, Austria, Oct. 2012.
43.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri and Hitoshi Nishijima : Aire controls organization of thymic microenvironment for the establishment of self tolerance, European Congress of Immunology 2012, Glasgow, Scotland, Sep. 2012.
44.
Mitsuru Matsumoto : Roles of Aire in thymic epithelial cells in the medulla for the establishment of self-tolerance, The Federation of Clinical Immunology Societies (FOCiS) 2012, Vancouver, Canada, Jun. 2012.
45.
Yasuhiro Mouri, Yumiko Nishikawa, Hitoshi Nishijima and Mitsuru Matsumoto : AIRE-dependent organization of thymic microenvironment for the establishment of self tolerance, 5th Congress of the Federation of Immunological Societies of Asia Oceania, New Delhi, India, Mar. 2012.
46.
Hitoshi Nishijima, Yasuhiro Mouri, Yumiko Nishikawa and Mitsuru Matsumoto : Cell-cycle dependent phosphorylation of AIRE protein exogenously expressed in HeLa cells, 4th International Conference on Drug Discovery & Therapy, Dubai, UAE, Feb. 2012.
47.
Mitsuru Matsumoto : Aire-dependent organization of thymic microenvironment for the establishment of self tolerance, The 4th symposium for the mext priority research on Immunological Self, Kyoto, Japan, Jan. 2012.
48.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri and Hitoshi Nishijima : Expression of Aire, a gene responsible for autoimmune disease, in ES cells, Cell Symposia: Stem Cell Programming & Re-programming, Lisbon, Portugal, Dec. 2011.
49.
Mitsuru Matsumoto, Yasuhiro Mouri, Yumiko Nishikawa and Hitoshi Nishijima : Aire-dependent organization of thymic microenvironment for the establishment of self tolerance, 5th Asian Congress on Autoimmunity, Suntec City, Singapore, Nov. 2011.
50.
Mitsuru Matsumoto, Yasuhiro Mouri, Yumiko Nishikawa and Hitoshi Nishijima : Role of self-antigen expressed by Aire-expressing cells in the negative selection process, 5th International Conference on Autoimmunity: Mechanisms and Novel Treatments, Hersonissos, Crete, Greece, Sep. 2011.
51.
Mitsuru Matsumoto, Yumiko Nishikawa, Yasuhiro Mouri and Hitoshi Nishijima : Aire-dependent organization of thymic microenvironment for the establishment of self tolerance, EUThyme-Rolduc Meeting, Leeuwenhorst, The Netherlands, May 2011.
52.
Yasuhiro Mouri, Masashi Yano, Miho Shinzawa, Yusuke Shimo, Yumiko Nishikawa, Koji Tamada, Lieping Chen, Josef M. Penninger, Jun-ichiro Inoue, Taishin Akiyama and Mitsuru Matsumoto : Lymphotoxin signal promotes thymic organogenesis by eliciting RANK expression in the embryonic thymic stroma, 13th International TNF Conference, TNF 2011, Awaji, Hyogo, May 2011.
53.
Mitsuru Matsumoto, Yumiko Nishikawa, Hitoshi Nishijima and Yasuhiro Mouri : Role of Aire in the organization of thymic microenvironment, Controversies in Rheumatology and Autoimmunity (CORA 2011), Florence, Italy, Mar. 2011.
54.
Yumiko Nishikawa, Yasuhiro Mouri, Hitoshi Nishijima and Mitsuru Matsumoto : Aire-dependent organization of thymic microenvironment for the establishment of self-tolerance, Cutting Edge Immunology and its Clinical Application, Hulshort, The Netherlands, Mar. 2011.
55.
Mitsuru Matsumoto : Roles of thymic epithelial cells for the establishment of self-tolerance, 14th International Congress of Immunology, Kobe, Aug. 2010.
56.
Yasuhiro Mouri, Yumiko Nishikawa and Mitsuru Matsumoto : Aire in thymic epithelial cells in the medulla controls developmental process of thymocytes, 14th International Congress of Immunology, Kobe, Aug. 2010.
57.
Yumiko Nishikawa, Yasuhiro Mouri and Mitsuru Matsumoto : Biphasic Aire expression in early embryos and in medullary thymic epithelial cells prior to end-stage terminal differentiation, 14th International Congress of Immunology, Kobe, Aug. 2010.
58.
Mitsuru Matsumoto and Yumiko Nishikawa : Expression of Aire, a histone-binding module for H3K4me0, in ES cell, Cold Spring Harbor Asia Conferences, Epigenetics, Chromatin & Transcription, Suzhou, China, May 2010.
59.
Yasuhiro Mouri and Mitsuru Matsumoto : Aire in thymic epithelial cells in the medulla controls developmental process of thymocytes, 7th International Congress on Autoimmunity, Ljubljana, Slovenia, May 2010.
60.
Mitsuru Matsumoto, Yumiko Nishikawa, Fumiko Hirota and Yasuhiro Mouri : AIRE and autoimmune disease, The 19th International Rheumatology Symposium, Kobe, Apr. 2010.
61.
Mitsuru Matsumoto, Yumiko Nishikawa, Fumiko Hirota, Masashi Yano, Hiroshi Kawamoto and Yasuhiro Mouri : Biphasic Aire expression in early embryos and in medullary thymic epithelial cells prior to end-stage terminal differentiation, ThymOz VI, Gladstone, Australia, Mar. 2010.
62.
Takashi Sakai, HoangNam Tran, Sun Mi Kim, Li Liu, Xichuan Teng, Yuji Shishido, Mukai-Sakai Rika, Mitsuru Matsumoto, Kazunori Ishimura, Yoshio Hayashi, Ryuji Kaji and Kiyoshi Fukui : Nucling, a novel stress-sensitive protein, regulates NF-kappa B activation, The 4th International Congress on Stress Responses in Biology and Medicine, The 4th Annual Meeting of the Biomedical Society for Stress Response, Sapporo, Oct. 2009.
63.
Mitsuru Matsumoto, Yumiko Nishikawa, Masashi Yano, Fumiko Hirota and Yasuhiro Mouri : Aire-dependent organization of thymic microenvironment, International KTCC 2009, Kyoto, Jun. 2009.
64.
Yasuhiro Mouri, Masashi Yano and Mitsuru Matsumoto : Roles of NIK in thymic epithelial cells for the establishment of central tolerance., 12th International TNF Conference, Madrid, Apr. 2009.
65.
Kazuko YORITA, Hideo Misaki, Mitsuru Matsumoto and Vincent Massey : Function of Two Arginine Residues of L-Lactate Oxidase, The 3rd International Conference on Oxygenases, Kyoto, Nov. 2000.
Proceeding of Domestic Conference:
1.
Minoru Matsumoto, Yasuyo Saijo, Mayuko Shimizu, Hirohisa Ogawa, Koichi Tsuneyama, Mitsuru Matsumoto and Takeshi Oya : 胸腺癌におけるAIRE発現と胸腺髄質上皮細胞との類似性, Proceedings of the Japanese Society of Pathology, Vol.111, No.1, 243, Apr. 2022.
Hitoshi Nishijima, Mizuki Sugita, Junko Morimoto, Minoru Matsumoto and Mitsuru Matsumoto : Functional and transcriptomic analysis of medullary thymic epithelial cells with augmented Aire expression, 第42回日本分子生物学会年会, Dec. 2019.
9.
Junko Morimoto, Hitoshi Nishijima, 松本 稔 and Mitsuru Matsumoto : Antigen Transfer from thymic epithelial cells to DCs contributes to the production of thymic Tregs., The 48th Annual meeting of the Japanese Society for Immunology, Dec. 2019.
Hitoshi Nishijima, Junko Morimoto, Minoru Matsumoto and Mitsuru Matsumoto : Transcriptomic analysis of medullary thymic epithelial cells with augmented Aire expression, 第47回日本免疫学会学術集会, Dec. 2018.
12.
Junko Morimoto, Hitoshi Nishijima, Minoru Matsumoto and Mitsuru Matsumoto : Analysis of the role of thymic APCs and Aire in the production of thymic Tregs, 第47回日本免疫学会学術集会, Dec. 2018.
Minoru Matsumoto, Hitoshi Nishijima, Junko Morimoto, Koichi Tsuneyama and Mitsuru Matsumoto : Transgenic human AIRE expression in NOD acquired resistance to the diabetes due to the impaired presentation of self-antigens in the pancreas, 第46回日本免疫学会学術集会, Dec. 2017.
Hitoshi Nishijima, Yoshiki Matsuoka, Yasuhiro Mouri, Junko Morimoto and Mitsuru Matsumoto : Possible defect in the Treg production in mice overexpressing AIRE/Aire which exhibit muscle-specific autoimmunity, 第45回日本免疫学会学術集会, Dec. 2016.
17.
Junko Morimoto, Yumiko Nishikawa, Hiroshi Kawano, Yasuhiro Mouri, Hitoshi Nishijima and Mitsuru Matsumoto : A novel Aire-dependent subset of medullary thymic epithelial cells that express Ly6 family protein, 第44回日本免疫学会学術集会, Nov. 2015.
18.
Hitoshi Nishijima, Yasuhiro Mouri, Junko Morimoto, Hiroshi Kawano and Mitsuru Matsumoto : Paradoxical development of polymyositis-like autoimmunity by the additive expression of AIRE, 第44回日本免疫学会学術集会, Nov. 2015.
19.
Hitoshi Nishijima, Junko Morimoto, Yasuhiro Mouri, Hiroshi Kawano, Koichi Ikuta and Mitsuru Matsumoto : Approaches to identify Aire-regulated non-tissue-restricted antigen genes by the ectopic expression of Aire in thymic cortex, 第43回日本免疫学会学術集会, Dec. 2014.
20.
Hiroshi Kawano, Junko Morimoto, Yasuhiro Mouri, Hitoshi Nishijima, Yasuhiko Nishioka and Mitsuru Matsumoto : Unexpected disturbance of the development of medullary thymic epithelial cells at immature stages by the long-term ablation of mature Aire-expressing cells, 第43回日本免疫学会学術集会, Dec. 2014.
21.
Hitoshi Nishijima, Kouichi Ikuta and Mitsuru Matsumoto : 胸腺皮質上皮細胞への異所性発現による Aire の機能解析, 24 Kyoto T cell Conference (KTCC), May 2014.
22.
Hitoshi Nishijima, Junko Morimoto, Yasuhiro Mouri, Yumiko Nishikawa, Kouichi Ikuta and Mitsuru Matsumoto : Requirement of Aire expression within thymic medulla but not cortex for establishing self-tolerance, 第42回日本免疫学会総会, Dec. 2013.
23.
Hitoshi Nishijima, Yasuhiro Mouri, Yumiko Nishikawa and Mitsuru Matsumoto : Cell-cycle dependent phosphorylation of AIRE protein exogenously expressed in HeLa cells, 第35回日本分子生物学会年会, Dec. 2012.
24.
Yasuhiro Mouri, Yumiko Nishikawa, Hitoshi Nishijima and Mitsuru Matsumoto : Differential requirement of Aire in two transgenic models for the negative selection of autoreactive thymocytes against self-antigens, 第41回日本免疫学会学術集会, Dec. 2012.
25.
Tomoyuki Yuasa, Toshiyuki Obata, Ichiro Yokota, Eiji Okamoto, Yoshiko Kanezaki, Hiroshi Maegawa, Fumiko Hirota, Kazuhiro Kishi, Seiichi Hashida, Hisao Nagaya, Ogura Yuko, Masuda Kazuhiko, Mitsuru Matsumoto, Toshio Matsumoto, Atsunori Kashiwagi and Yousuke Ebina : Soluble insulin receptor ectodomain is elevated in the plasma of patients with diabetes., International Symposium on Diabetes - Kickoff of Hannover-Tokushima Research Communiation, Mar. 2009.
26.
Takashi Sakai, Li Liu, Rika Sakai, Ryuji Kaji, Tasuku Mitani, Mitsuru Matsumoto, Kazunori Toida, Yuji Shishido and Kiyoshi Fukui : Nucling is important for the upregulation of Apaf-1/procaspase-9cytochrome c apoptosome following cellular stress in vivo, The 77th Annual Meeting of the Japanese Biochemical Society, Oct. 2004.
Et cetera, Workshop:
1.
Junko Morimoto and Mitsuru Matsumoto : Aire欠損mTECは異所性CTLA-4を発現することによって胸腺内DCの機能に影響を及ぼす., 第30回KTCC, Oct. 2021.
2.
Junko Morimoto and Mitsuru Matsumoto : 胸腺髄質上皮細胞から樹状細胞への抗原転移におけるAireの役割, 日本胸腺研究会 2021年2月 on line 開催, Feb. 2021.
Analysis of two homologous proteins Ly6C1 and Ly6C2 on immune homeostasis (Project/Area Number: 19K07626 )
Elucidation of the pathogenesis of autoimmune diseases based on AIRE and development of novel therapies for the autoimmune diseases (Project/Area Number: 19H03699 )
Studies on the mechanisms underlying autoimmune ambivalence (Project/Area Number: 18K19564 )
Mechanisms for the creation of neo-self in the thymus (Project/Area Number: 16H06496 )
Comprehensive understanding of neo-self generation, function, and structure (Project/Area Number: 16H06495 )
International support for the reseach activities for the production, function and structure of neo-self (Project/Area Number: 16K21731 )
Elucidation of the pathogenesis of autoimmune disease through the functional study of AIRE (Project/Area Number: 16H05342 )
Studies on the mechanisms for autoimmune phenotype in disease model mouse (Project/Area Number: 26461464 )
Expression of Ly6C/6G defines a novel Aire-dependent subset of medullary thymic epithelial cells with tolerogenic function (Project/Area Number: 26460577 )
Role of Aire in the early development of zebrafish (Project/Area Number: 25670231 )
Studies aiming the establishing self-tolerance for the cure of autoimmune disease (Project/Area Number: 25293223 )
Functional analysis of the non-HLA genes within major histocompatibility complex (MHC) region with the use of gene-targeted mice (Project/Area Number: 13670320 )
Functional analysis of the non-HLA genes within major histocompatibility complex (MHC) region with the use of gene-targeted mice (Project/Area Number: 11557013 )