Network


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

Hotspot


Dive into the research topics where Masashi Kanayama is active.

Publication


Featured researches published by Masashi Kanayama.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Interleukin-17A Deficiency Accelerates Unstable Atherosclerotic Plaque Formation in Apolipoprotein E-Deficient Mice

Keiko Danzaki; Yutaka Matsui; Masahiro Ikesue; Daichi Ohta; Koyu Ito; Masashi Kanayama; Daisuke Kurotaki; Junko Morimoto; Yoichiro Iwakura; Hideo Yagita; Hiroyuki Tsutsui; Toshimitsu Uede

Objective—Interleukin(IL)-17A, an inflammatory cytokine, has been implicated in atherosclerosis, in which inflammatory cells within atherosclerotic plaques express IL-17A. However, its role in the development of atheroscelrosis remains to be controversial. Methods and Results—To directly examine the role of IL-17A in atherosclerosis, we generated apolipoprotein E (ApoE)/IL-17A double-deficient (ApoE−/−IL-17A−/−) mice. Mice were fed with high-fat diet (HFD) for either 8 or 16 weeks, both starting at ages of 6 to 8 weeks. We found that splenic CD4+ T-cells produced high amounts of IL-17A in ApoE−/− mice after HFD feeding for 8 weeks. Atherosclerosis was significantly accelerated in HFD-fed ApoE−/−IL-17A−/− mice compared with ApoE−/− mice. Splenic CD4+ T-cells of ApoE−/−IL-17A−/− mice after HFD feeding for 8 weeks, but not for 16 weeks, exhibited increased interferon gamma and decreased IL-5 production. Importantly, formation of vulnerable plaque as evidenced by reduced numbers of vascular smooth muscle cells and reduced type I collagen deposition in the plaque was detected in ApoE−/−IL-17A−/− mice after HFD feeding for 8 weeks. Conclusion—These results suggest that IL-17A regulates the early phase of atherosclerosis development after HFD feeding and plaque stability, at least partly if not all by modulating interferon gamma and IL-5 production from CD4+ T-cells.


Journal of Immunology | 2009

α9 Integrin and Its Ligands Constitute Critical Joint Microenvironments for Development of Autoimmune Arthritis

Masashi Kanayama; Daisuke Kurotaki; Junko Morimoto; Asano T; Yutaka Matsui; Yosuke Nakayama; Yoshinari Saito; Koyu Ito; Chiemi Kimura; Norimasa Iwasaki; Koji Suzuki; Tanenobu Harada; Hong Mei Li; Jun Uehara; Tadaaki Miyazaki; Akio Minami; Shigeyuki Kon; Toshimitsu Uede

Osteopontin is critically involved in rheumatoid arthritis; however, the molecular cross-talk between osteopontin and joint cell components that leads to the inflammatory joint destruction is largely unknown. We found that not only osteopontin but also tenascin-C and their common receptor, α9 integrin, are expressed at arthritic joints. The local production of osteopontin and tenascin-C is mainly due to synovial fibroblasts and, to a lesser extent, synovial macrophages. Synovial fibroblasts and macrophages express α9 integrin, and autocrine and paracrine interactions of α9 integrin on synovial fibroblasts and macrophages and its ligands contribute differently to the production of proinflammatory cytokines and chemokines. α9 integrin is also involved in the recruitment and accumulation of inflammatory cells. Inhibition of α9 integrin function with an anti-α9 integrin Ab significantly reduces the production of arthrogenic cytokines and chemokines and ameliorates ongoing arthritis. Thus, we identified α9 integrin as a critical intrinsic regulator that controls the development of autoimmune arthritis.


Matrix Biology | 2009

The differential amino acid requirement within osteopontin in α4 and α9 integrin-mediated cell binding and migration

Koyu Ito; Shigeyuki Kon; Yosuke Nakayama; Daisuke Kurotaki; Yoshinari Saito; Masashi Kanayama; Chiemi Kimura; Hongyan Diao; Junko Morimoto; Yutaka Matsui; Toshimitsu Uede

Osteopontin (OPN) contains at least two major integrin recognition domains, Arg159-Gly-Asp161 (RGD) and Ser162-Val-Val-Tyr-Gly-Leu-Arg168 (SVVYGLR), recognized by alphavbeta3 and alpha5beta1 and alpha4 and alpha9 integrins, respectively. OPN is specifically cleaved by thrombin and matrix metalloproteinase (MMP)-3 or MMP-7 at a position of Arg168/Ser169 (R/S) and Gly166/Leu167 (G/L), respectively. We in this study examined the requirement of residues within SVVYGLR for the alpha4 and alpha9 integrin recognition and how MMP-cleavage influences the integrin recognition. The residues, Val164, Tyr165, and Leu167 are critical for alpha4 and alpha9 integrin recognition in both cell adhesion and cell migration. The residue Arg168 is additionally required for alpha9 integrin recognition in cell adhesion and this explains why alpha9 integrin binds to only thrombin cleaved form of OPN. alpha4 integrin is able to bind to SVVYG (MMP-cleaved form of RAA OPN-N half), while alpha9 integrin is not, supporting the above notion that Arg168 is additionally required for alpha9 integrin-mediated cell adhesion. The residue Val163 is important for alpha4, but not for alpha9 integrin recognition in cell migration. Importantly, we found that the replacement of Arg168 by Ala (R168A mutant) induces the augmentation of cell migration via alpha4 and alpha9 integrins.


Journal of Immunology | 2011

α9β1 Integrin-Mediated Signaling Serves as an Intrinsic Regulator of Pathogenic Th17 Cell Generation

Masashi Kanayama; Junko Morimoto; Yutaka Matsui; Masahiro Ikesue; Keiko Danzaki; Daisuke Kurotaki; Koyu Ito; Toshimichi Yoshida; Toshimitsu Uede

The interaction between matricellular proteins such as tenascin-C (TN-C) and osteopontin (OPN) and integrins has been implicated in the pathology of rheumatoid arthritis in which Th17 cells are recognized as primary pathogenic cells. The differentiation of Th17 cells is tightly regulated by cytokines derived from APCs, receiving various signals including TLR stimuli. In this study, we used a collagen-induced arthritis model and found that increased numbers of α9 integrin-positive conventional dendritic cells and macrophage were detectable in the draining lymph node (dLN) shortly following first immunization, and these cells produced both TN-C and OPN, ligands for α9 integrin. α9 integrin-mediated signaling, induced by TN-C and OPN, promoted the production of Th17-related cytokines by conventional dendritic cells and macrophages in synergy with TLR2 and 4 signaling. This led to the Th17 cell differentiation and arthritis development. Moreover, Th17 cells generated under blocking of α9 integrin-mediated signaling showed low level of CCR6 expression and impaired migration ability toward CCL20. Thus, we have identified α9 integrin-mediated signaling by TN-C and OPN as a novel intrinsic regulator of pathogenic Th17 cell generation that contributes to the development of rheumatoid arthritis.


Journal of Immunology | 2011

CSF-1–Dependent Red Pulp Macrophages Regulate CD4 T Cell Responses

Daisuke Kurotaki; Shigeyuki Kon; Kyeonghwa Bae; Koyu Ito; Yutaka Matsui; Yosuke Nakayama; Masashi Kanayama; Chiemi Kimura; Yoshinori Narita; Takashi Nishimura; Kazuya Iwabuchi; Matthias Mack; Nico van Rooijen; Shimon Sakaguchi; Toshimitsu Uede; Junko Morimoto

The balance between immune activation and suppression must be regulated to maintain immune homeostasis. Tissue macrophages (MΦs) constitute the major cellular subsets of APCs within the body; however, how and what types of resident MΦs are involved in the regulation of immune homeostasis in the peripheral lymphoid tissues are poorly understood. Splenic red pulp MΦ (RPMs) remove self-Ags, such as blood-borne particulates and aged erythrocytes, from the blood. Although many scattered T cells exist in the red pulp of the spleen, little attention has been given to how RPMs prevent harmful T cell immune responses against self-Ags. In this study, we found that murine splenic F4/80hiMac-1low MΦs residing in the red pulp showed different expression patterns of surface markers compared with F4/80+Mac-1hi monocytes/MΦs. Studies with purified cell populations demonstrated that F4/80hiMac-1low MΦs regulated CD4+ T cell responses by producing soluble suppressive factors, including TGF-β and IL-10. Moreover, F4/80hiMac-1low MΦs induced the differentiation of naive CD4+ T cells into functional Foxp3+ regulatory T cells. Additionally, we found that the differentiation of F4/80hiMac-1low MΦs was critically regulated by CSF-1, and in vitro-generated bone marrow-derived MΦs induced by CSF-1 suppressed CD4+ T cell responses and induced the generation of Foxp3+ regulatory T cells in vivo. These results suggested that splenic CSF-1–dependent F4/80hiMac-1low MΦs are a subpopulation of RPMs and regulate peripheral immune homeostasis.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2011

Syndecan-4 Deficiency Limits Neointimal Formation After Vascular Injury by Regulating Vascular Smooth Muscle Cell Proliferation and Vascular Progenitor Cell Mobilization

Masahiro Ikesue; Yutaka Matsui; Daichi Ohta; Keiko Danzaki; Koyu Ito; Masashi Kanayama; Daisuke Kurotaki; Junko Morimoto; Tetsuhito Kojima; Hiroyuki Tsutsui; Toshimitsu Uede

Objective—Syndecan-4 (Syn4) is a heparan sulfate proteoglycan and works as a coreceptor for various growth factors. We examined whether Syn4 could be involved in the development of neointimal formation in vivo. Methods and Results—Wild-type (WT) and Syn4-deficient (Syn4−/−) mice were subjected to wire-induced femoral artery injury. Syn4 mRNA was upregulated after vascular injury in WT mice. Neointimal formation was attenuated in Syn4−/− mice, concomitantly with the reduction of Ki67-positive vascular smooth muscle cells (VSMCs). Basic-fibroblast growth factor– or platelet-derived growth factor-BB–induced proliferation, extracellular signal-regulated kinase activation, and expression of cyclin D1 and Bcl-2 were impaired in VSMCs from Syn4−/− mice. To examine the role of Syn4 in bone marrow (BM)–derived vascular progenitor cells (VPCs) and vascular walls, we generated chimeric mice by replacing the BM cells of WT and Syn4−/− mice with those of WT or Syn4−/− mice. Syn4 expressed by both vascular walls and VPCs contributed to the neointimal formation after vascular injury. Although the numbers of VPCs were compatible between WT and Syn4−/− mice, mobilization of VPCs from BM after vascular injury was defective in Syn4−/− mice. Conclusion—Syn4 deficiency limits neointimal formation after vascular injury by regulating VSMC proliferation and VPC mobilization. Therefore, Syn4 may be a novel therapeutic target for preventing arterial restenosis after angioplasty.


PLOS ONE | 2012

Ganglioside GM3 has an essential role in the pathogenesis and progression of rheumatoid arthritis.

Yukinori Tsukuda; Norimasa Iwasaki; Naoki Seito; Masashi Kanayama; Naoki Fujitani; Yasuro Shinohara; Yasuhiko Kasahara; Tomohiro Onodera; Koji Suzuki; Asano T; Akio Minami; Tadashi Yamashita

Rheumatoid arthritis (RA), a chronic systemic inflammatory disorder that principally attacks synovial joints, afflicts over 2 million people in the United States. Interleukin (IL)-17 is considered to be a master cytokine in chronic, destructive arthritis. Levels of the ganglioside GM3, one of the most primitive glycosphingolipids containing a sialic acid in the structure, are remarkably decreased in the synovium of patients with RA. Based on the increased cytokine secretions observed in in vitro experiments, GM3 might have an immunologic role. Here, to clarify the association between RA and GM3, we established a collagen-induced arthritis mouse model using the null mutation of the ganglioside GM3 synthase gene. GM3 deficiency exacerbated inflammatory arthritis in the mouse model of RA. In addition, disrupting GM3 induced T cell activation in vivo and promoted overproduction of the cytokines involved in RA. In contrast, the amount of the GM3 synthase gene transcript in the synovium was higher in patients with RA than in those with osteoarthritis. These findings indicate a crucial role for GM3 in the pathogenesis and progression of RA. Control of glycosphingolipids such as GM3 might therefore provide a novel therapeutic strategy for RA.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Integrin α9 on lymphatic endothelial cells regulates lymphocyte egress

Koyu Ito; Junko Morimoto; Akio Kihara; Yutaka Matsui; Daisuke Kurotaki; Masashi Kanayama; Szandor Simmons; Masaru Ishii; Dean Sheppard; Akinori Takaoka; Toshimitsu Uede

Significance The lymphatic system plays critical roles in immune surveillance by providing a route for circulating immune cells. α9 integrin is a signature molecule of lymphatic endothelial cells (LECs), and has been suggested to have a significant role in lymphatic functions. Here we found that in draining lymph nodes (LNs) of mice treated with complete Freund adjuvant, blockade of α9 integrin induced deficiency of lymphocytes in medullary and cortical sinuses, which is the characteristic feature of impaired lymphocyte egress from LNs. In addition, we found that stimulation of embryo-derived LECs with the α9 ligand tenascin-C induced secretion of sphingosine 1-phosphate, which is the critical factor for lymphocyte egress. Thus, we suggest a possible role of α9 integrin-dependent lymphocyte egress from inflamed LNs. Sphingosine 1-phosphate (S1P) plays a role in lymphocyte egress from lymphoid organs. However, it remains unclear how S1P production and secretion are regulated. We show that under inflammatory conditions, α9 integrin, which is closely associated with activated β1 integrin, and its ligand, tenascin-C, colocalize on medullary and cortical sinuses of draining lymph nodes (dLNs), which is a gate for lymphocyte exit, and that inhibition of lymphocyte egress is evident by blockade of α9 integrin-mediated signaling at dLNs. Based on in vitro analysis using lymphatic endothelial cells obtained from mice embryos, we suggested the possibility that stimulation of lymphatic endothelial cells by tenascin-C enhances S1P secretion in an α9 integrin-dependent manner without affecting S1P synthesis and/or degradation. Blockade of α9 integrin-mediated signaling reduced lymphocyte egress from dLNs in several models, including experimental autoimmune encephalomyelitis, where it improved clinical scores and pathology. Therefore, manipulating α9 integrin function may offer a therapeutic strategy for treating various inflammatory disorders.


Rheumatology | 2014

α9β1 integrin acts as a critical intrinsic regulator of human rheumatoid arthritis

Asano T; Norimasa Iwasaki; Shigeyuki Kon; Masashi Kanayama; Junko Morimoto; Akio Minami; Toshimitsu Uede

OBJECTIVE The role of the joint tissue microenvironment in the pathogenesis of human RA has recently attracted much attention. The present study investigated the roles of α9β1 integrin and its ligands in synovial specimens of human RA patients in generating the unique human arthritic tissue microenvironment. METHODS Synovial fibroblasts and macrophages were isolated from the synovial tissue of patients with RA or OA. The expression of α9β1 integrin was analysed using FACS with multicolour staining. The production of MMPs and proinflammatory cytokines was analysed in cultures of synovial fibroblasts and macrophages with α9β1 integrin ligands. RESULTS Synovial fibroblasts and macrophages derived from arthritic joints spontaneously secreted tenascin-C and osteopontin. Synovial fibroblasts and macrophages obtained from patients with RA expressed α9β1 integrins, a common receptor for osteopontin and tenascin-C. In the synovial fibroblasts of RA, the amount of tenascin-C protein produced was much greater than that of osteopontin in synovial fibroblasts of RA. Importantly, autocrine and paracrine interactions of α9β1 integrin and tenascin-C induced the expression of MMPs and IL-6 in synovial fibroblasts, as well as TNF-α and IL-1β in synovial macrophages. CONCLUSION These findings indicate that autocrine and paracrine interaction of α9β1 integrin and tenascin-C in the joint tissue microenvironment contributes to the pathogenesis of RA. Therefore α9β1 integrin may become a potential therapeutic target for RA.


PLOS ONE | 2014

A novel cryptic binding motif, LRSKSRSFQVSDEQY, in the C-terminal fragment of MMP-3/7-cleaved osteopontin as a novel ligand for α9β1 integrin is involved in the anti-type II collagen antibody-induced arthritis.

Shigeyuki Kon; Yosuke Nakayama; Naoki Matsumoto; Koyu Ito; Masashi Kanayama; Chiemi Kimura; Hitomi Kouro; Dai Ashitomi; Tadashi Matsuda; Toshimitsu Uede

Osteopontin (OPN) is a multifunctional protein that has been linked to various intractable inflammatory diseases. One way by which OPN induces inflammation is the production of various functional fragments by enzyme cleavage. It has been well appreciated that OPN is cleaved by thrombin, and/or matrix metalloproteinase-3 and -7 (MMP-3/7). Although the function of thrombin-cleaved OPN is well characterized, little is known about the function of MMP-3/7-cleaved OPN. In this study, we found a novel motif, LRSKSRSFQVSDEQY, in the C-terminal fragment of MMP-3/7-cleaved mouse OPN binds to α9β1 integrin. Importantly, this novel motif is involved in the development of anti-type II collagen antibody-induced arthritis (CAIA). This study provides the first in vitro and in vivo evidence that OPN cleavage by MMP-3/7 is an important regulatory mechanism for CAIA.

Collaboration


Dive into the Masashi Kanayama's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toshimitsu Uede

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Toshimitsu Uede

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge