Network


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

Hotspot


Dive into the research topics where Keiichi Iwanami is active.

Publication


Featured researches published by Keiichi Iwanami.


Arthritis & Rheumatism | 2008

Crucial role of the interleukin-6/interleukin-17 cytokine axis in the induction of arthritis by glucose-6-phosphate isomerase

Keiichi Iwanami; Isao Matsumoto; Yoko Tanaka-Watanabe; Asuka Inoue; Masahiko Mihara; Yoshiyuki Ohsugi; Mizuko Mamura; Daisuke Goto; Satoshi Ito; Akito Tsutsumi; Tadamitsu Kishimoto; Takayuki Sumida

OBJECTIVE To clarify the glucose-6-phosphate isomerase (GPI)-specific CD4+ T cell lineage involved in GPI-induced arthritis and to investigate their pathologic and regulatory roles in the induction of the disease. METHODS DBA/1 mice were immunized with GPI to induce arthritis. CD4+ T cells and antigen-presenting cells were cocultured with GPI, and cytokines in the supernatant were analyzed by enzyme-linked immunosorbent assay. Anti-interferon-gamma (anti-IFNgamma) monoclonal antibody (mAb), anti-interleukin-17 (anti-IL-17) mAb, or the murine IL-6 receptor (IL-6R) mAb MR16-1 was injected at different time points, and arthritis development was monitored visually. After MR16-1 was injected, percentages of Th1, Th2, Th17, and Treg cells were analyzed by flow cytometry, and CD4+ T cell proliferation was analyzed using carboxyfluorescein diacetate succinimidyl ester. RESULTS GPI-specific CD4+ T cells were found to be differentiated to Th1 and Th17 cells, but not Th2 cells. Administration of anti-IL-17 mAb on day 7 significantly ameliorated arthritis (P < 0.01), whereas administration of anti-IFNgamma mAb exacerbated arthritis. Neither anti-IL-17 mAb nor anti-IFNgamma mAb administration on day 14 ameliorated arthritis. Administration of MR16-1 on day 0 or day 3 protected against arthritis induction, and MR16-1 administration on day 8 significantly ameliorated existing arthritis (P < 0.05). After administration of MR16-1, there was marked suppression of Th17 differentiation, without an increase in Th1, Th2, or Treg cells, and CD4+ T cell proliferation was also suppressed. CONCLUSION IL-6 and Th17 play an essential role in GPI-induced arthritis. Since it has previously been shown that treatment with a humanized anti-IL-6R mAb has excellent effects in patients with rheumatoid arthritis (RA), we propose that the IL-6/IL-17 axis might also be involved in the generation of RA, especially in the early effector phase.


Arthritis Research & Therapy | 2008

Therapeutic effects of antibodies to tumor necrosis factor-α, interleukin-6 and cytotoxic T-lymphocyte antigen 4 immunoglobulin in mice with glucose-6-phosphate isomerase induced arthritis

Isao Matsumoto; Hua Zhang; Takanori Yasukochi; Keiichi Iwanami; Yoko Tanaka; Asuka Inoue; Daisuke Goto; Satoshi Ito; Akito Tsutsumi; Takayuki Sumida

IntroductionImmunization with glucose-6-phosphate isomerase (GPI) induces severe arthritis in DBA/1 mice. The present study was designed to identify the cytokines and co-stimulatory molecules involved in the development of GPI-induced arthritis.MethodsArthritis was induced in DBA/1 mice with 300 μg human recombinant GPI. CD4+ T cells and antigen-presenting cells from splenocytes of arthritic mice were cultured in the presence of GPI. Tumor necrosis factor (TNF)-α, IFN-γ, IL-2, IL-4, IL-5, IL-6, IL-10, and IL-12 levels were assessed using cytometric bead array. Monoclonal antibodies to TNF-α, IFN-γ, IL-12, CD40L, inducible co-stimulator (ICOS), and cytotoxic T-lymphocyte antigen 4 immunoglobulin (CTLA-4Ig) were used to block TNF-α and IFN-γ production, examine clinical index in mice with GPI-induced arthritis, and determine anti-GPI antibody production.ResultsLarge amounts of TNF-α and IFN-γ and small amounts of IL-2 and IL-6 were produced by splenocytes from mice with GPI-induced arthritis. Anti-TNF-α mAbs and CTLA-4Ig suppressed TNF-α production, whereas anti-IFN-γ mAbs, anti-IL-12 mAbs, and CTLA-4 Ig inhibited IFN-γ production. A single injection of anti-TNF-α and anti-IL-6 mAbs and two injections of CTLA-4Ig reduced the severity of arthritis in mice, whereas injections of anti-IFN-γ and anti-IL-12 mAbs tended to exacerbate arthritis. Therapeutic efficacy tended to correlate with reduction in anti-GPI antibodies.ConclusionTNF-α and IL-6 play an important role in GPI-induced arthritis, whereas IFN-γ appears to function as a regulator of arthritis. Because the therapeutic effects of the tested molecules used in this study are similar to those in patients with rheumatoid arthritis, GPI-induced arthritis appears to be a suitable tool with which to examine the effect of various therapies on rheumatoid arthritis.


Arthritis Research & Therapy | 2008

Arthritogenic T cell epitope in glucose-6-phosphate isomerase-induced arthritis

Keiichi Iwanami; Isao Matsumoto; Yoko Tanaka; Asuka Inoue; Daisuke Goto; Satoshi Ito; Akito Tsutsumi; Takayuki Sumida

IntroductionArthritis induced by immunisation with glucose-6-phosphate isomerase (GPI) in DBA/1 mice was proven to be T helper (Th) 17 dependent. We undertook this study to identify GPI-specific T cell epitopes in DBA/1 mice (H-2q) and investigate the mechanisms of arthritis generation.MethodsFor epitope mapping, the binding motif of the major histocompatibility complex (MHC) class II (I-Aq) from DBA/1 mice was identified from the amino acid sequence of T cell epitopes and candidate peptides of T cell epitopes in GPI-induced arthritis were synthesised. Human GPI-primed CD4+ T cells and antigen-presenting cells (APCs) were co-cultured with each synthetic peptide and the cytokine production was measured by ELISA to identify the major epitopes. Synthetic peptides were immunised in DBA/1 mice to investigate whether arthritis could be induced by peptides. After immunisation with the major epitope, anti-interleukin (IL) 17 monoclonal antibody (mAb) was injected to monitor arthritis score. To investigate the mechanisms of arthritis induced by a major epitope, cross-reactivity to mouse GPI peptide was analysed by flow cytometry and anti-GPI antibodies were measured by ELISA. Deposition of anti-GPI antibodies on the cartilage surface was detected by immunohistology.ResultsWe selected 32 types of peptides as core sequences from the human GPI 558 amino acid sequence, which binds the binding motif, and synthesised 25 kinds of 20-mer peptides for screening, each containing the core sequence at its centre. By epitope mapping, human GPI325–339 was found to induce interferon (IFN) γ and IL-17 production most prominently. Immunisation with human GPI325–339 could induce polyarthritis similar to arthritis induced by human GPI protein, and administration of anti-IL-17 mAb significantly ameliorated arthritis (p < 0.01). Th17 cells primed with human GPI325–339 cross-reacted with mouse GPI325–339, and led B cells to produce anti-mouse GPI antibodies, which were deposited on cartilage surface.ConclusionsHuman GPI325–339 was identified as a major epitope in GPI-induced arthritis, and proved to have the potential to induce polyarthritis. Understanding the pathological mechanism of arthritis induced by an immune reaction to a single short peptide could help elucidate the pathogenic mechanisms of autoimmune arthritis.


Annals of the Rheumatic Diseases | 2007

Visfatin (pre-B cell colony-enhancing factor) gene expression in patients with rheumatoid arthritis

H Matsui; Akito Tsutsumi; Makoto Sugihara; T. Suzuki; Keiichi Iwanami; Mika Kohno; Daisuke Goto; Isao Matsumoto; Satoshi Ito; Takayuki Sumida

Visfatin, also known as pre-B cell colony enhancing factor, is a cytokine-like protein originally cloned from peripheral blood mononuclear cells (PBMC) as a factor that enhances differentiation of pre-B cells in synergy with interleukin 7 (IL-)7 and stem cell factor.1 Visfatin is mainly produced in visceral white adipose tissues, and shows an insulin-like effect by binding to insulin receptors.2 Generally, visfatin is regarded as an adipokine, and elevated levels of serum visfatin in patients with type 2 diabetes,3 and obesity,4 have been reported. Interestingly, in visceral adipose tissues, macrophages seem to be the major source …


Modern Rheumatology | 2008

Use of laser microdissection in the analysis of renal-infiltrating T cells in MRL/lpr mice

Yingge Wang; Satoshi Ito; Yusuke Chino; Keiichi Iwanami; Takanori Yasukochi; Daisuke Goto; Isao Matsumoto; Taichi Hayashi; Kazuhiko Uchida; Takayuki Sumida

To clarify the role of T cells in the kidneys of MRL/MpJ-lpr (MRL/lpr) mice, cytokine mRNA expression was analyzed, and tissue localization of T cells was examined by immunohistochemistry. Cells infiltrating the glomeruli, glomerular circumference, and perivascular areas in ten female MRL/lpr mice were captured by laser microdissection (LMD). Nested reverse transcription polymerase chain reaction (RT-PCR) of samples was performed with primers specific for β-actin, T-cell receptor β chain (TCR-Cβ), Thy-1, B220, CD4, CD8, interleukin (IL)-2, IL-4, IL-10, IL-13, IL-17, and interferon (IFN)-γ. Frozen sections of lesions were also stained immunohistochemically. B220, MAC-1, Thy-1, CD4, and CD8 staining was observed in glomeruli and perivascular areas, especially in glomerular circumference areas. T cells infiltrating the glomeruli, glomerular circumference areas, and perivascular areas produce INF-γ, IL-13, and IL-17 predominately. IL-10 positivity was identified in 60% of perivascular T cells but not in a substantial number of glomerular or periglomerular T cells. The results of our study suggest that the pathogenesis of renal lesions in MRL/lpr mice is complex and not due simply to the Th1 and Th2 balance. These findings also support the concept of different molecular mechanisms for glomerulonephritis and vasculitis in these mice.


Arthritis Research & Therapy | 2009

Tumor necrosis factor α-induced adipose-related protein expression in experimental arthritis and in rheumatoid arthritis

Asuka Inoue; Isao Matsumoto; Yoko Tanaka; Keiichi Iwanami; Akihiro Kanamori; Naoyuki Ochiai; Daisuke Goto; Satoshi Ito; Takayuki Sumida

IntroductionTumor necrosis factor-alpha (TNFα) plays a pivotal role in rheumatoid arthritis (RA); however, the mechanism of action of TNFα antagonists in RA is poorly defined. Immunization of DBA/1 mice with glucose-6-phosphate isomerase (GPI) induces severe acute arthritis. This arthritis can be controlled by TNFα antagonists, suggesting similar etiology to RA. In this study, we explored TNFα-related mechanisms of arthritis.MethodsFirst, we performed GeneChip analysis using splenocytes of mice with GPI-induced arthritis. Expression of TNFα-induced adipose-related protein (TIARP) mRNA and protein in spleens, joints and lymph nodes was evaluated, and fluctuation of TIARP mRNA was analyzed after administration of anti-TNFα monoclonal antibody (mAb). Localization of TIARP in spleen and joints was also explored. Six-transmembrane epithelial antigen of the prostate (STEAP) families of proteins, the human ortholog of TIARP gene, were also evaluated in human peripheral blood mononucleocytes and synovium.ResultsAmong the arrayed TNFα-related genes, the expression of TIARP mRNA was the highest (more than 20 times the control). TIARP mRNA was detected specifically in joints and spleens of arthritic mice, and their levels in the synovia correlated with severity of joint swelling. Treatment with anti-TNF mAb significantly reduced TIARP mRNA expression in splenocytes. Among the splenocytes, CD11b+ cells were the main source of TIARP mRNA. Immunohistochemistry showed that TIARP protein was mainly localized in hyperplastic synovium. Among the STEAP family of proteins, STEAP4 was highly upregulated in joints of patients with RA and especially co-localized with CD68+ macrophages.ConclusionsThe results shed light on the new mechanism of action of TNFα antagonists in autoimmune arthritis, suggesting that TIARP plays an important role in inflammatory arthritis, through the regulation of inflammatory cytokines.


Modern Rheumatology | 2012

Six-transmembrane epithelial antigen of prostate4 (STEAP4) is a tumor necrosis factor alpha-induced protein that regulates IL-6, IL-8, and cell proliferation in synovium from patients with rheumatoid arthritis

Yoko Tanaka; Isao Matsumoto; Keiichi Iwanami; Asuka Inoue; Reiko Minami; Naoto Umeda; Akihiro Kanamori; Naoyuki Ochiai; Keiji Miyazawa; Makoto Sugihara; Taichi Hayashi; Daisuke Goto; Satoshi Ito; Takayuki Sumida

Human six-transmembrane epithelial antigen of prostate4 (STEAP4), an ortholog of mouse tumor necrosis factor-α-induced adipose-related protein (TIARP), plays a role in tumor necrosis factor (TNF)-dependent arthritis models. However, its role in rheumatoid arthritis (RA) is still obscure. This study explored such a role for STEAP4. The expressions of STEAP4, TNFα, and IL-6 were compared in synovia of RA and osteoarthritis patients. STEAP4 induction was examined in TNFα-stimulated fibroblast-like synoviocytes (FLS) in vitro. FLS (with/without TNFα stimulation) were also analyzed for IL-6 expression after STEAP4 knockdown, using siRNA or transfection with STEAP4-plasmid DNA. IL-8, cell proliferation, and apoptosis were also evaluated in STEAP4-overexpressing FLS. The expression of STEAP4 in joints correlated with TNFα expression, specifically in RA synovium. In the cultured FLS, STEAP4 protein expression was augmented by TNFα activation, and localized in endosomal/lysosomal compartments. STEAP4 downregulation by siRNA enhanced the expression of IL-6 mRNA, while STEAP4 overexpression suppressed IL-6 and IL-8 expression, inhibited cell proliferation, and induced apoptosis via caspase-3. The results indicated that human STEAP4 is regulated by TNFα in synovium, where it controls IL-6 secretion and proliferation of FLS, suggesting that STEAP4 might potentially suppress the pathogenesis of TNFα-induced arthritis such as RA.


Clinical and Experimental Immunology | 2009

B cells play a crucial role as antigen-presenting cells and collaborate with inflammatory cytokines in glucose-6-phosphate isomerase-induced arthritis

Y. Tanaka-Watanabe; Isao Matsumoto; Keiichi Iwanami; Asuka Inoue; Daisuke Goto; Satoshi Ito; Akito Tsutsumi; Takayuki Sumida

Anti‐glucose‐6‐phosphate isomerase (GPI) antibodies from K/BxN mice directly induce arthritis; however, the transfer of these antibodies from mice with GPI‐induced arthritis does not induce arthritis. CD4+ T cells play an important role in the induction and effector phase in this model; however, the roles of B cells and immunoglobulins (Igs) have not been elucidated. We investigated the roles of B cells and Igs in GPI‐induced arthritis by using adoptive transfer system into SCID mice. Transfer of splenocytes of male DBA/1 mice immunized with GPI into SCID mice induced arthritis on day 6 in the latter, in association with the production of anti‐GPI antibodies. Co‐localization of C3 and IgG on the articular surface was identified in arthritic SCID mice. Inoculation of IgG (or anti‐GPI antibodies) and CD19+‐depleted splenocytes from arthritic DBA/1 mice induced arthritis in SCID mice, but not CD19+‐depleted or CD4+‐depleted splenocytes from DBA/1 mice. In vitro analysis of cytokine production by splenocytes from DBA/1 arthritic mice demonstrated production of large amounts of tumour necrosis factor (TNF)‐α and interleukin (IL)‐6 in an antigen‐specific manner (P < 0·01), and production was dominated by CD19+‐depleted than CD4+‐depleted splenocytes (P < 0·05). Addition of IgG from DBA/1 arthritic mice to the culture enhanced TNF‐α but not IL‐6 production, and this effect was blocked by anti‐Fcγ receptor antibody. In vivo analysis of neutralization with TNF‐α protected arthritis completely in SCID mice. Our results highlight the important role of B cells in GPI‐induced arthritis as autoantibody producers, and these autoantibodies can trigger joint inflammation in orchestration with inflammatory cytokines, especially TNF‐α.


Arthritis Research & Therapy | 2009

Altered peptide ligands inhibit arthritis induced by glucose-6-phosphate isomerase peptide

Keiichi Iwanami; Isao Matsumoto; Yohei Yoshiga; Asuka Inoue; Yuya Kondo; Kayo Yamamoto; Yoko Tanaka; Reiko Minami; Taichi Hayashi; Daisuke Goto; Satoshi Ito; Yasuharu Nishimura; Takayuki Sumida

IntroductionImmunosuppressants, including anti-TNFα antibodies, have remarkable effects in rheumatoid arthritis; however, they increase infectious events. The present study was designed to examine the effects and immunological change of action of altered peptide ligands (APLs) on glucose-6-phosphate isomerase (GPI) peptide-induced arthritis.MethodsDBA/1 mice were immunized with hGPI325-339, and cells of draining lymph node (DLN) were stimulated with hGPI325-339 to investigate the T-cell receptor (TCR) repertoire of antigen-specific CD4+ T cells by flow cytometry. Twenty types of APLs with one amino acid substitution at a TCR contact site of hGPI325-339 were synthesized. CD4+ T cells primed with human GPI and antigen-presenting cells were co-cultured with each APL and cytokine production was measured by ELISA to identify antagonistic APLs. Antagonistic APLs were co-immunized with hGPI325-339 to investigate whether arthritis could be antigen-specifically inhibited by APL. After co-immunization, DLN cells were stimulated with hGPI325-339 or APL to investigate Th17 and regulatory T-cell population by flow cytometry, and anti-mouse GPI antibodies were measured by ELISA.ResultsHuman GPI325-339-specific Th17 cells showed predominant usage of TCRVβ8.1 8.2. Among the 20 synthesized APLs, four (APL 6; N329S, APL 7; N329T, APL 12; G332A, APL 13; G332V) significantly reduced IL-17 production by CD4+ T cells in the presence of hGPI325-339. Co-immunization with each antagonistic APL markedly prevented the development of arthritis, especially APL 13 (G332V). Although co-immunization with APL did not affect the population of Th17 and regulatory T cells, the titers of anti-mouse GPI antibodies in mice co-immunized with APL were significantly lower than in those without APL.ConclusionsWe prepared antagonistic APLs that antigen-specifically inhibited the development of experimental arthritis. Understanding the inhibitory mechanisms of APLs may pave the way for the development of novel therapies for arthritis induced by autoimmune responses to ubiquitous antigens.


PLOS ONE | 2012

Activation of Invariant NKT Cells with Glycolipid Ligand α-Galactosylceramide Ameliorates Glucose-6-Phosphate Isomerase Peptide-Induced Arthritis

Masanobu Horikoshi; Daisuke Goto; Seiji Segawa; Yohei Yoshiga; Keiichi Iwanami; Asuka Inoue; Yuki Tanaka; Isao Matsumoto; Takayuki Sumida

Objective Invariant natural killer T (iNKT) cells regulate collagen-induced arthritis (CIA) when activated by their potent glycolipid ligand, alpha-galactosylceramide (α-GalCer). Glucose-6-phosphate isomerase (GPI)-induced arthritis is a closer model of human rheumatoid arthritis based on its association with CD4+ T cells and cytokines such as TNF-α and IL-6 than CIA. Dominant T cell epitope peptide of GPI (GPI325-339) can induce arthritis similar to GPI-induced arthritis. In this study, we investigated the roles of activation of iNKT cells by α-GalCer in GPI peptide-induced arthritis. Methods Arthritis was induced in susceptible DBA1 mice with GPI peptide and its severity was assessed clinically. The arthritic mice were treated with either the vehicle (DMSO) or α-GalCer. iNKT cells were detected in draining lymph nodes (dLNs) by flow cytometry, while serum anti-GPI antibody levels were measured by enzyme-linked immunosorbent assay. To evaluate GPI peptide-specific cytokine production from CD4+ T cells, immunized mice were euthanized and dLN CD4+ cells were re-stimulated by GPI-peptide in the presence of antigen-presenting cells. Results α-GalCer induced iNKT cell expansion in dLNs and significantly decreased the severity of GPI peptide-induced arthritis. In α-GalCer-treated mice, anti-GPI antibody production (total IgG, IgG1, IgG2b) and IL-17, IFN-γ, IL-2, and TNF-α produced by GPI peptide-specific T cells were significantly suppressed at day 10. Moreover, GPI-reactive T cells from mice immunized with GPI and α-GalCer did not generate any cytokines even when these cells were co-cultured with APC from mice immunized with GPI alone. In vitro depletion of iNKT cells did not alter the suppressive effect of α-GalCer on CD4+ T cells. Conclusion α-GalCer significantly suppressed GPI peptide-induced arthritis through the suppression of GPI-specific CD4+ T cells.

Collaboration


Dive into the Keiichi Iwanami's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge