Network


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

Hotspot


Dive into the research topics where Nobuaki Mizutani is active.

Publication


Featured researches published by Nobuaki Mizutani.


Journal of Immunology | 2014

IL-17A Promotes the Exacerbation of IL-33–Induced Airway Hyperresponsiveness by Enhancing Neutrophilic Inflammation via CXCR2 Signaling in Mice

Nobuaki Mizutani; Takeshi Nabe; Shin Yoshino

Neutrophilic airway inflammation is a hallmark of patients with severe asthma. Although we have reported that both IL-33 and IL-17A contributed to IgE-mediated neutrophilic inflammation in mice, the relationship remains unclear. In this article, we examined how IL-17A modifies IL-33–induced neutrophilic inflammation and airway hyperresponsiveness (AHR). IL-33 was intratracheally administered to BALB/c mice on days 0–2; furthermore, on day 7, the effect of the combination of IL-33 and IL-17A was evaluated. Compared with IL-33 or IL-17A alone, the combination exacerbated neutrophilic inflammation and AHR, associated with more increased levels of lung glutamic acid-leucine-arginine+ CXC chemokines, including CXCL1, CXCL2, and CXCL5, and infiltration by alveolar macrophages expressing CXCR2. Treatment with anti-CXCR2 mAb or depletion of alveolar macrophages repressed neutrophilic inflammation and AHR; in addition, depletion of neutrophils suppressed AHR. These findings prompted us to examine the role of CXCR2 in IgE-sensitized mice; a single treatment with anti-CXCR2 mAb in the seventh Ag challenge inhibited late-phase airway obstruction, AHR, and neutrophilic inflammation. In addition to inhibition, multiple treatments during the fourth to seventh challenge attenuated early-phase airway obstruction, eosinophilic inflammation, and goblet cell hyperplasia associated with the reduction of Th2 cytokine production, including IL-4, IL-5, and IL-13. Collectively, IL-33 cooperated with IL-17A to exacerbate AHR by enhancing neutrophilic inflammation via CXCR2 signaling; furthermore, CXCR2 signaling derived Th2 responses. We thus suggest the underlying mechanisms of IL-33 and IL-17A in allergic asthma and CXCR2 as potential therapeutic targets for the disease.


Immunology | 2013

Interleukin-33 and alveolar macrophages contribute to the mechanisms underlying the exacerbation of IgE-mediated airway inflammation and remodelling in mice.

Nobuaki Mizutani; Takeshi Nabe; Shin Yoshino

Allergen‐specific IgE has long been regarded as a major molecular component of allergic asthma. Additionally, there is increasing evidence of the important roles of interleukin‐33 (IL‐33) in the disease. Here, we show that IL‐33 and alveolar macrophages play essential roles in the exacerbation of IgE‐mediated airway inflammation and remodelling. BALB/c mice passively sensitized with ovalbumin (OVA)‐specific IgE monoclonal antibody (mAb) were challenged with OVA seven times intratracheally. The seventh challenge exacerbated airway inflammation and remodelling compared with the fourth challenge; furthermore, markedly increased expression of IL‐33 in the lungs was observed at the fourth and seventh challenges. When anti‐IL‐33 or anti‐ST2 antibody was administered during the fourth to seventh challenge, airway inflammation and remodelling were significantly inhibited at the seventh challenge. Because increases of IL‐33+ and ST2+ alveolar macrophages and ST2+ CD4+ T cells in the lungs were observed at the fourth challenge, the roles of macrophages and CD4+ cells were investigated. Depletion of macrophages by 2‐chloroadenosine during the fourth to seventh challenge suppressed airway inflammation and remodelling, and IL‐33 production in the lung at the seventh challenge; additionally, anti‐CD4 mAb inhibited airway inflammation, but not airway remodelling and IL‐33 production. Meanwhile, treatment with 2‐chloroadenosine or anti‐CD4 mAb decreased IL‐33‐induced airway inflammation in normal mice; airway remodelling was repressed only by 2‐chloroadenosine. These results illustrate that macrophage‐derived IL‐33 contributes to the exacerbation of IgE‐mediated airway inflammation by mechanisms associated with macrophages and CD4+ cells, and airway remodelling through the activation of macrophages.


European Journal of Pharmacology | 2015

Production of interleukin (IL)-33 in the lungs during multiple antigen challenge-induced airway inflammation in mice, and its modulation by a glucocorticoid.

Takeshi Nabe; Hiroki Wakamori; Chihiro Yano; Ayumi Nishiguchi; Rino Yuasa; Hitomi Kido; Yusaku Tomiyama; Ayumi Tomoda; Haruka Kida; Anna Takiguchi; Masaya Matsuda; Keiichi Ishihara; Satoshi Akiba; Susumu Ohya; Hiroyuki Fukui; Nobuaki Mizutani; Shin Yoshino

Although interleukin (IL)-33 is a candidate aggravator of asthma, the cellular sources of IL-33 in the lungs during the progression of antigen-induced airway inflammation remain unclear. Furthermore, it has not been determined whether the antigen-induced production of IL-33 can be pharmacologically modulated in vivo. In this study, we examined the production of IL-33 in the lungs of sensitized mice during multiple intratracheal challenges with the antigen, ovalbumin. The 1st challenge clearly induced the IL-33 production in the lungs, and it was enhanced by the 2nd-4th challenges. IL-33 mRNA transcription was also induced after these challenges. An immunohistochemical analysis revealed that the cellular sources of IL-33 after the 1st challenge were mainly bronchial epithelial cells, while those after the 3rd challenge were not only the epithelial cells, but also inflammatory cells that infiltrated the lungs. Flow cytometric analyses indicated that approximately 20% and 10% of the IL-33-producing cells in the lungs were M2 macrophages and conventional dendritic cells, respectively. A systemic treatment with dexamethasone before the 1st challenge potently suppressed the IL-33 production. When dexamethasone was administered before the respective challenges, production of the IL-33 protein and the infiltration of IL-33-producing M2 macrophages and dendritic cells into the lungs in the 3rd challenge were also suppressed. In conclusion, the cellular sources of IL-33 in the lungs were dynamically altered during multiple challenges: not only bronchial epithelial cells, but also the M2 macrophages and dendritic cells that infiltrated the lungs produced IL-33. The production of IL-33 was susceptible to the glucocorticoid treatment.


British Journal of Pharmacology | 2013

Roles of basophils and mast cells infiltrating the lung by multiple antigen challenges in asthmatic responses of mice

Takeshi Nabe; Kouki Matsuya; K. Akamizu; M. Fujita; T. Nakagawa; M. Shioe; Haruka Kida; Anna Takiguchi; Hiroki Wakamori; Masanori Fujii; Keiichi Ishihara; Satoshi Akiba; Nobuaki Mizutani; Shin Yoshino; Dd D. Chaplin

Mast cell hyperplasia has been observed in the lungs of mice with experimental asthma, but few reports have studied basophils. Here, we attempted to discriminate and quantify mast cells and basophils in the lungs in a murine asthma model, determine if both cells were increased by multiple antigen challenges and assess the roles of those cells in asthmatic responses.


Immunology | 2015

Thymic stromal lymphopoietin‐induced interleukin‐17A is involved in the development of IgE‐mediated atopic dermatitis‐like skin lesions in mice

Nobuaki Mizutani; Chutha Sae-Wong; Sureeporn Kangsanant; Takeshi Nabe; Shin Yoshino

Atopic dermatitis (AD) is a chronic inflammatory skin disease associated with elevated levels of allergen‐specific IgE. Although thymic stromal lymphopoietin (TSLP) and interleukin‐17A (IL‐17A) have been considered as important factors in allergic diseases, their relationships in AD have not been fully defined. Here, we show the contribution of TSLP‐induced IL‐17A responses to IgE‐mediated AD‐like skin lesions. BALB/c mice passively sensitized by intraperitoneal injections of ovalbumin (OVA)‐specific IgE monoclonal antibody (mAb) were challenged with OVA applied to the skin six times. Treatment with anti‐TSLP mAb during the second to sixth challenges inhibited IgE‐mediated AD‐like skin lesions and IL‐17A production in lymph nodes. Furthermore, the increased number of IL‐17A‐producing CD4+ and γδ T cells in lymph nodes and neutrophilic inflammation in the skin were reduced by anti‐TSLP mAb. These findings prompted us to examine the roles of IL‐17A. Treatment with anti‐IL‐17A mAb suppressed the AD‐like skin lesions and neutrophilic inflammation; anti‐Gr‐1 mAb also inhibited them. Furthermore, treatment with CXCR2 antagonist reduced the AD‐like skin lesions and neutrophilic inflammation accompanied by the reduction of IL‐17A production; the increased CXCR2 expression in the epidermal cells was suppressed by anti‐TSLP mAb. Meanwhile, these treatments, except for anti‐Gr‐1 mAb, inhibited the increased mast cell accumulation in the skin. Collectively, the mechanism of IgE mediating IL‐17A‐producing CD4+ and γδ T cells through TSLP by repeated antigen challenges is involved in AD‐like skin lesions associated with skin inflammation, such as neutrophil and mast cell accumulation; TSLP may regulate CXCR2 signalling‐induced IL‐17A production.


Immunology | 2014

Intratracheal exposure to Fab fragments of an allergen‐specific monoclonal antibody regulates asthmatic responses in mice

Shin Yoshino; Nobuaki Mizutani; Daiko Matsuoka; Chutha Sae-Wong

Fab fragments (Fabs) maintain the ability to bind to specific antigens but lack effector functions due to the absence of the Fc portion. In the present study, we tested whether Fabs of an allergen‐specific monoclonal antibody (mAb) were able to regulate asthmatic responses in mice. Asthmatic responses were induced in BALB/c mice by passive sensitization with anti‐ovalbumin (OVA) polyclonal antibodies (pAbs) (day 0) and by active sensitization with OVA (days 0 and 14), followed by intratracheal (i.t.) challenge with OVA on day 1 and days 28, 29, 30 and 35. Fabs prepared by the digestion of an anti‐OVA IgG1 (O1‐10) mAb with papain were i.t. administered only once 30 min before antigenic challenge on day 1 or day 35. The results showed that i.t. administration of O1‐10 Fabs with OVA markedly suppressed the early and/or late phases of asthmatic responses caused by passive and active sensitization. Similar results were obtained when Fabs of anti‐OVA IgG2b mAb (O2B‐3) were i.t. administered. In contrast, neither i.t. injection of intact 01‐10/O2B‐3 nor systemic injection of O1‐10 Fabs suppressed the asthmatic responses. In vitro studies revealed that the capture of OVA by O1‐10 Fabs prevented the subsequent binding of intact anti‐OVA pAbs to the captured OVA. These results suggest that asthmatic responses may be down‐regulated by the i.t. exposure to Fabs of an allergen‐specific mAb via a mechanism involving the capture of allergen by Fabs in the respiratory tract before the interaction of intact antibody and allergen essential for the induction of asthmatic responses.


Immunology Letters | 2014

Allergen-specific regulation of allergic rhinitis in mice by intranasal exposure to IgG1 monoclonal antibody Fab fragments against pathogenic allergen

Daiko Matsuoka; Nobuaki Mizutani; Chutha Sae-Wong; Shin Yoshino

Fab fragments (Fabs) have the ability to bind to specific antigens but lack the Fc portion for binding to receptors on immune and inflammatory cells that play a critical role in allergic diseases. In the present study, we investigated whether Fabs of an allergen-specific IgG1 monoclonal antibody (mAb) inhibited allergic rhinitis in mice. BALB/c mice sensitized by intraperitoneal injections of ovalbumin (OVA) plus alum on days 0 and 14 were intranasally challenged with OVA on days 28-30, and 35. Fabs prepared by the digestion of an anti-OVA IgG1 mAb (O1-10) with papain were also intranasally administered 15min before each OVA challenge. The results showed that treatment with O1-10 Fabs significantly suppressed the sneezing frequency, associated with decrease of OVA-specific IgE in the serum and infiltration by mast cells in the nasal mucosa seen following the fourth antigenic challenge; additionally, the level of mouse mast cell protease-1, a marker of mast cell activation, in serum was decreased. Furthermore, infiltration of eosinophils and goblet cell hyperplasia in the nasal mucosa at the fourth challenge were inhibited by treatment with O1-10 Fabs. In conclusion, these results suggest that intranasal exposure to Fabs of a pathogenic antigen-specific IgG1 mAb may be effective in regulating allergic rhinitis through allergen capture by Fabs in the nasal mucosa before the interaction of the intact antibody and allergen.


European Journal of Pharmacology | 2015

Semaphorin 7A plays a critical role in IgE-mediated airway inflammation in mice

Nobuaki Mizutani; Takeshi Nabe; Shin Yoshino

Elevated allergen-specific IgE levels are a hallmark of allergic asthma, a disease involving chronic airway inflammation characterized by airway hyperresponsiveness (AHR); neutrophilic airway inflammation is found in patients with severe asthma. Furthermore, we have reported that interleukin (IL)-33 and IL-17A contribute to IgE-mediated AHR through neutrophilic inflammation in mice. Meanwhile, semaphorins regulating neuronal and immune function have been focused on in several diseases. Here, we investigated whether semaphorin 7A (SEMA7A) is related to IgE-mediated neutrophilic inflammation in mice. BALB/c mice sensitized with antigen-specific IgE monoclonal antibody were repeatedly challenged by the antigen. When anti-SEMA7A antibody was administered during the fourth to seventh challenges, the infiltration by macrophages, lymphocytes, neutrophils, and eosinophils in the lungs was reduced at the seventh challenge (P<0.05, 0.05, 0.01, and 0.05, respectively). However, the increased production of IL-4, IL-5, IL-13, IL-33, IL-17A, IL-6, and CXCL1 in the lungs was not suppressed. In histological analysis, the epithelial cells, blood vessels, and inflammatory cells in the lungs of IgE-sensitized mice showed SEMA7A expression; plexin C1 for the receptor was expressed in the inflammatory cells. Meanwhile, we examined the effect of anti-SEMA7A antibody on AHR and neutrophilic inflammation enhanced by the collaborative action of IL-33 and IL-17A in normal mice, resulting in the suppression of these responses (P<0.05 and 0.01, respectively). Collectively, we demonstrated that SEMA7A plays a critical role in IgE-mediated neutrophilic airway inflammation. Therefore, SEMA7A may be a potential therapeutic target for severe allergic asthma showing neutrophilic airway inflammation.


European Journal of Pharmacology | 2016

Topical skin treatment with Fab fragments of an allergen-specific IgG1 monoclonal antibody suppresses allergen-induced atopic dermatitis-like skin lesions in mice.

Chutha Sae-Wong; Nobuaki Mizutani; Sureeporn Kangsanant; Shin Yoshino

Fab fragments (Fabs), which lack effector functions due to the absence of the Fc portion, maintain the ability to bind to specific allergens. In the present study, we examined whether Fabs of an allergen-specific IgG1 monoclonal antibody (mAb) were able to regulate allergen-induced atopic dermatitis-like skin lesions in mice. BALB/c mice passively sensitized with ovalbumin (OVA)-specific IgE mAb were repeatedly challenged with OVA applied to the skin after sodium dodecyl sulfate treatment. Fabs prepared by the digestion of anti-OVA IgG1 mAb (O1-10) with papain were applied to the skin 30min before the OVA challenges followed by measurement of clinical symptoms including erythema/hemorrhage, edema, scarring/dryness, and excoriation/erosion of the skin. Treatment with O1-10 Fabs, but not intact O1-10, showed inhibition of clinical symptoms (P<0.01) induced by the repeated OVA challenges in the sensitized mice; O1-10 Fabs suppressed histological changes such as epidermal hyperplasia (P<0.01) and the accumulation of mast cells (P<0.01) and neutrophils (P<0.01). Furthermore, treatment with O1-10 Fabs inhibited the increase in levels of IL-13 (P<0.01) and IL-17A production (P<0.05) in the lymph nodes of the sensitized mice. Additionally, the increased level of OVA in serum following the repeated OVA challenges in the sensitized mice was reduced by the treatment (P<0.05). These results suggest that topical application of pathogenic allergen-specific IgG1 mAb Fabs to the skin of mice is effective in suppressing allergen-induced atopic dermatitis-like skin lesions, suggesting that allergen-specific mAb Fabs could be used as a tool to regulate allergen-induced atopic dermatitis.


British Journal of Pharmacology | 2016

Intranasal exposure to monoclonal antibody Fab fragments to Japanese cedar pollen Cry j1 suppresses Japanese cedar pollen‐induced allergic rhinitis

Shin Yoshino; Nobuaki Mizutani

Fab fragments (Fabs) of antibodies have the ability to bind to specific allergens but lack the Fc portion that exerts effector functions via binding to receptors including FcεR1 on mast cells. In the present study, we investigated whether intranasal administration of the effector function‐lacking Fabs of a monoclonal antibody IgG1 (mAb, P1‐8) to the major allergen Cry j1 of Japanese cedar pollen (JCP) suppressed JCP‐induced allergic rhinitis in mice.

Collaboration


Dive into the Nobuaki Mizutani's collaboration.

Top Co-Authors

Avatar

Shin Yoshino

Kobe Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chutha Sae-Wong

Prince of Songkla University

View shared research outputs
Top Co-Authors

Avatar

Anna Takiguchi

Kyoto Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Daiko Matsuoka

Kobe Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Haruka Kida

Kyoto Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Hiroki Wakamori

Kyoto Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Keiichi Ishihara

Kyoto Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Satoshi Akiba

Kyoto Pharmaceutical University

View shared research outputs
Top Co-Authors

Avatar

Sureeporn Kangsanant

Kobe Pharmaceutical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge