Network


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

Hotspot


Dive into the research topics where Keiko Nakano is active.

Publication


Featured researches published by Keiko Nakano.


The Journal of Allergy and Clinical Immunology | 2012

IL-35 production by inducible costimulator (ICOS)–positive regulatory T cells reverses established IL-17–dependent allergic airways disease

Gregory S. Whitehead; Rhonda H. Wilson; Keiko Nakano; Lauranell H. Burch; Hideki Nakano; Donald N. Cook

BACKGROUND Recent evidence suggests that IL-17 contributes to airway hyperresponsiveness (AHR); however, the mechanisms that suppress the production of this cytokine remain poorly defined. OBJECTIVE We sought to identify the regulatory cells and molecules that suppress IL-17-dependent allergic airways disease. METHODS Mice were sensitized by means of airway instillations of ovalbumin together with low levels of LPS. Leukocyte recruitment to the lung and AHR were assessed after daily challenges with aerosolized ovalbumin. Flow cytometry, quantitative PCR, and gene-targeted mice were used to identify naturally arising subsets of regulatory T (Treg) cells and their cytokines required for the suppression of established allergic airway disease. RESULTS Allergic sensitization through the airway primed both effector and regulatory responses. Effector responses were initially dominant and led to airway inflammation and IL-17-dependent AHR. However, after multiple daily allergen challenges, IL-17 production and AHR decreased, even though pulmonary levels of T(H)17 cells remained high. This loss of AHR was reversible and required the expansion of a Treg cell subset expressing both forkhead box protein 3 and inducible costimulator. These Treg cells also expressed the regulatory cytokines IL-10, TGF-β, and IL-35. Whereas IL-10 and TGF-β were dispensable for suppression of AHR, IL-35 was required. CONCLUSION IL-35 production by inducible costimulator-positive Treg cells can suppress IL-17 production and thereby reverse established, IL-17-dependent AHR in mice. Targeting this pathway might therefore be of therapeutic value for treating allergic asthma in human subjects.


Mucosal Immunology | 2012

Pulmonary CD103(+) dendritic cells prime Th2 responses to inhaled allergens.

Hideki Nakano; M E Free; Gregory S. Whitehead; Shuichiro Maruoka; Rhonda H. Wilson; Keiko Nakano; Donald N. Cook

Allergic asthma stems largely from the actions of T helper 2 (Th2) cells, but the pathways that initiate Th2 responses to inhaled allergens are not fully understood. In the lung, there are two major subsets of dendritic cells (DCs), displaying CD11b or CD103. We found that after taking up inhaled ovalbumin in vivo, purified CD103+ DCs from the lung or lung-draining lymph nodes primed Th2 differentiation ex vivo. Th2 induction by CD103+ DCs was also seen when cockroach or house dust mite allergens were used. In contrast, CD11bhi DCs primed Th1 differentiation. Moreover, mice lacking CD103+ DCs displayed diminished Th2 priming to various inhaled allergens and did not develop asthma-like responses following subsequent allergen challenge. Low-level antigen presentation by CD103+ DCs was necessary, but not sufficient for Th2 priming. Together, these findings show that CD103+ DCs have a significant role in priming Th2 responses to inhaled allergens.


The Journal of Allergy and Clinical Immunology | 2012

Mechanisms of allergy and clinical immunologyIL-35 production by inducible costimulator (ICOS)–positive regulatory T cells reverses established IL-17–dependent allergic airways disease

Gregory S. Whitehead; Rhonda H. Wilson; Keiko Nakano; Lauranell H. Burch; Hideki Nakano; Donald N. Cook

BACKGROUND Recent evidence suggests that IL-17 contributes to airway hyperresponsiveness (AHR); however, the mechanisms that suppress the production of this cytokine remain poorly defined. OBJECTIVE We sought to identify the regulatory cells and molecules that suppress IL-17-dependent allergic airways disease. METHODS Mice were sensitized by means of airway instillations of ovalbumin together with low levels of LPS. Leukocyte recruitment to the lung and AHR were assessed after daily challenges with aerosolized ovalbumin. Flow cytometry, quantitative PCR, and gene-targeted mice were used to identify naturally arising subsets of regulatory T (Treg) cells and their cytokines required for the suppression of established allergic airway disease. RESULTS Allergic sensitization through the airway primed both effector and regulatory responses. Effector responses were initially dominant and led to airway inflammation and IL-17-dependent AHR. However, after multiple daily allergen challenges, IL-17 production and AHR decreased, even though pulmonary levels of T(H)17 cells remained high. This loss of AHR was reversible and required the expansion of a Treg cell subset expressing both forkhead box protein 3 and inducible costimulator. These Treg cells also expressed the regulatory cytokines IL-10, TGF-β, and IL-35. Whereas IL-10 and TGF-β were dispensable for suppression of AHR, IL-35 was required. CONCLUSION IL-35 production by inducible costimulator-positive Treg cells can suppress IL-17 production and thereby reverse established, IL-17-dependent AHR in mice. Targeting this pathway might therefore be of therapeutic value for treating allergic asthma in human subjects.


Mucosal Immunology | 2013

Migratory properties of pulmonary dendritic cells are determined by their developmental lineage.

Hideki Nakano; J E Burgents; Keiko Nakano; Gregory S. Whitehead; C Cheong; C D Bortner; Donald N. Cook

The chemokine receptor, CCR7, directs the migration of dendritic cells (DCs) from peripheral tissue to draining lymph nodes (LNs). However, it is unknown whether all pulmonary DCs possess migratory potential. Using novel Ccr7gfp reporter mice, we found that Ccr7 is expressed in CD103+ and a CD14med/lo subset of CD11bhi classical (c)DCs but not in monocyte-derived (mo)DCs, including Ly-6ChiCD11bhi inflammatory DCs and CD14hiCD11bhi DCs. Consequently, cDCs migrated to lung-draining LNs but moDCs did not. Mice lacking the chemokine receptor, CCR2, also lacked inflammatory DCs in the lung after lipopolysaccharide inhalation but retained normal levels of migratory DCs. Conversely, the lungs of fms-like tyrosine kinase 3 ligand (Flt3L)-deficient mice lacked cDCs but retained moDCs, which were functionally mature but did not express Ccr7 and were uniformly non-migratory. Thus, the migratory properties of pulmonary DCs are determined by their developmental lineage.


Journal of Immunology | 2015

Complement Receptor C5aR1/CD88 and Dipeptidyl Peptidase-4/CD26 Define Distinct Hematopoietic Lineages of Dendritic Cells

Hideki Nakano; Timothy P. Moran; Keiko Nakano; Kevin Gerrish; Carl D. Bortner; Donald N. Cook

Differential display of the integrins CD103 and CD11b are widely used to distinguish two major dendritic cell (DC) subsets in nonlymphoid tissues. CD103+ DCs arise from FLT3-dependent DC precursors (preDCs), whereas CD11bhi DCs can arise either from preDCs or FLT3-independent monocytes. Functional characterization of these two lineages of CD11bhi DCs has been hindered by the lack of a widely applicable method to distinguish between them. We performed gene expression analysis of fractionated lung DCs from C57BL/6 mice and found that monocyte-derived DCs (moDCs), including CD11bhiLy-6Clo tissue-resident and CD11bhiLy-6Chi inflammatory moDCs, express the complement 5a receptor 1/CD88, whereas preDC-derived conventional DCs (cDCs), including CD103+ and CD11bhi cDCs, express dipeptidyl peptidase-4/CD26. Flow cytometric analysis of multiple organs, including the kidney, liver, lung, lymph nodes, small intestine, and spleen, confirmed that reciprocal display of CD88 and CD26 can reliably distinguish FLT3-independent moDCs from FLT3-dependent cDCs in C57BL/6 mice. Similar results were obtained when DCs from BALB/c mice were analyzed. Using this novel approach to study DCs in mediastinal lymph nodes, we observed that most blood-derived lymph node–resident DCs, as well as tissue-derived migratory DCs, are cDCs. Furthermore, cDCs, but not moDCs, stimulated naive T cell proliferation. We anticipate that the use of Abs against CD88 and CD26 to distinguish moDCs and cDCs in multiple organs and mouse strains will facilitate studies aimed at assigning specific functions to distinct DC lineages in immune responses.


Mucosal Immunology | 2015

Trif -dependent induction of Th17 immunity by lung dendritic cells

B J Hsia; Gregory S. Whitehead; S Y Thomas; Keiko Nakano; Kymberly M. Gowdy; J J Aloor; Hideki Nakano; Donald N. Cook

Allergic asthma is thought to stem largely from maladaptive T helper 2 (Th2) responses to inhaled allergens, which in turn lead to airway eosinophilia and airway hyperresponsiveness (AHR). However, many individuals with asthma have airway inflammation that is predominantly neutrophilic and resistant to treatment with inhaled glucocorticoids. An improved understanding of the molecular basis of this form of asthma might lead to improved strategies for its treatment. Here, we identify novel roles of the adaptor protein, TRIF (TIR-domain-containing adapter-inducing interferon-β), in neutrophilic responses to inhaled allergens. In different mouse models of asthma, Trif-deficient animals had marked reductions in interleukin (IL)-17, airway neutrophils, and AHR compared with wild-type (WT) mice, whereas airway eosinophils were generally similar in these two strains. Compared with lung dendritic cells (DCs) from WT mice, lung DCs from Trif-deficient mice displayed impaired lipopolysaccharide (LPS)-induced migration to regional lymph nodes, lower levels of the costimulatory molecule, CD40, and produced smaller amounts of the T helper 17 (Th17)-promoting cytokines, IL-6, and IL-1β. When cultured with allergen-specific, naive T cells, Trif-deficient lung DCs stimulated robust Th2 cell differentiation but very weak Th1 and Th17 cell differentiation. Together, these findings reveal a TRIF-CD40-Th17 axis in the development of IL-17-associated neutrophilic asthma.


Journal of Leukocyte Biology | 2017

Distinct functions of CXCR4, CCR2, and CX3CR1 direct dendritic cell precursors from the bone marrow to the lung

Hideki Nakano; Miranda R. Lyons-Cohen; Gregory S. Whitehead; Keiko Nakano; Donald N. Cook

Precursors of dendritic cells (pre‐DCs) arise in the bone marrow (BM), egress to the blood, and finally migrate to peripheral tissue, where they differentiate to conventional dendritic cells (cDCs). Upon their activation, antigen‐bearing cDCs migrate from peripheral tissue to regional lymph nodes (LNs) in a manner dependent on the chemokine receptor, CCR7. To maintain immune homeostasis, these departing cDCs must be replenished by new cDCs that develop from pre‐DCs, but the molecular signals that direct pre‐DC trafficking from the BM to the blood and peripheral tissues remain poorly understood. In the present study, we found that pre‐DCs express the chemokine receptors CXCR4, CCR2, and CX3CR1, and that each of these receptors has a distinct role in pre‐DC trafficking. Flow cytometric analysis of pre‐DCs lacking CXCR4 revealed that this receptor is required for the retention of pre‐DCs in the BM. Analyses of mice lacking CCR2 or CX3CR1, or both, revealed that they promote pre‐DC migration to the lung at steady state. CCR2, but not CX3CR1, was required for pre‐DC migration to the inflamed lung. Thus, these multiple chemokine receptors cooperate in a step‐wise fashion to coordinate the trafficking of pre‐DCs from the BM to the circulation and peripheral tissues.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2015

Inhaled house dust programs pulmonary dendritic cells to promote type 2 T-cell responses by an indirect mechanism

X Timothy P. Moran; Keiko Nakano; Gregory S. Whitehead; Seddon Y. Thomas; Donald N. Cook; Hideki Nakano

The induction of allergen-specific T helper 2 (Th2) cells by lung dendritic cells (DCs) is a critical step in allergic asthma development. Airway delivery of purified allergens or microbial products can promote Th2 priming by lung DCs, but how environmentally relevant quantities and combinations of these factors affect lung DC function is unclear. Here, we investigated the ability of house dust extract (HDE), which contains a mixture of environmental adjuvants, to prime Th2 responses against an innocuous inhaled antigen. Inhalational exposure to HDE conditioned lung conventional DCs, but not monocyte-derived DCs, to induce antigen-specific Th2 differentiation. Conditioning of DCs by HDE was independent of Toll-like receptor 4 signaling, indicating that environmental endotoxin is dispensable for programming DCs to induce Th2 responses. DCs directly treated with HDE underwent maturation but were poor stimulators of Th2 differentiation. In contrast, DCs treated with bronchoalveolar lavage fluid (BALF) from HDE-exposed mice induced robust Th2 differentiation. DC conditioning by BALF was independent of the proallergic cytokines IL-25, IL-33, and thymic stromal lymphopoietin. BALF treatment of DCs resulted in upregulation of CD80 but low expression of CD40, CD86, and IL-12p40, which was associated with Th2 induction. These findings support a model whereby environmental adjuvants in house dust indirectly program DCs to prime Th2 responses by triggering the release of endogenous soluble factor(s) by airway cells. Identifying these factors could lead to novel therapeutic targets for allergic asthma.


Journal of Clinical Investigation | 2017

TNF is required for TLR ligand–mediated but not protease-mediated allergic airway inflammation

Gregory S. Whitehead; Seddon Y. Thomas; Karim H Shalaby; Keiko Nakano; Timothy P. Moran; James M. Ward; Gordon P. Flake; Hideki Nakano; Donald N. Cook

Asthma is associated with exposure to a wide variety of allergens and adjuvants. The extent to which overlap exists between the cellular and molecular mechanisms triggered by these various agents is poorly understood, but it might explain the differential responsiveness of patients to specific therapies. In particular, it is unclear why some, but not all, patients benefit from blockade of TNF. Here, we characterized signaling pathways triggered by distinct types of adjuvants during allergic sensitization. Mice sensitized to an innocuous protein using TLR ligands or house dust extracts as adjuvants developed mixed eosinophilic and neutrophilic airway inflammation and airway hyperresponsiveness (AHR) following allergen challenge, whereas mice sensitized using proteases as adjuvants developed predominantly eosinophilic inflammation and AHR. TLR ligands, but not proteases, induced TNF during allergic sensitization. TNF signaled through airway epithelial cells to reprogram them and promote Th2, but not Th17, development in lymph nodes. TNF was also required during the allergen challenge phase for neutrophilic and eosinophilic inflammation. In contrast, TNF was dispensable for allergic airway disease in a protease-mediated model of asthma. These findings might help to explain why TNF blockade improves lung function in only some patients with asthma.


Mucosal Immunology | 2018

MyD88-dependent dendritic and epithelial cell crosstalk orchestrates immune responses to allergens

S Y Thomas; Gregory S. Whitehead; M Takaku; J M Ward; X Xu; Keiko Nakano; M R Lyons-Cohen; Hideki Nakano; Kymberly M. Gowdy; P A Wade; Donald N. Cook

Sensitization to inhaled allergens is dependent on activation of conventional dendritic cells (cDCs) and on the adaptor molecule, MyD88. However, many cell types in the lung express Myd88, and it is unclear how signaling in these different cell types reprograms cDCs and leads to allergic inflammation of the airway. By combining ATAC-seq with RNA profiling, we found that MyD88 signaling in cDCs maintained open chromatin at select loci even at steady state, allowing genes to be rapidly induced during allergic sensitization. A distinct set of genes related to metabolism was indirectly controlled in cDCs through MyD88 signaling in airway epithelial cells (ECs). In mouse models of asthma, Myd88 expression in ECs was critical for eosinophilic inflammation, whereas Myd88 expression in cDCs was required for Th17 cell differentiation and consequent airway neutrophilia. Thus, both cell-intrinsic and cell-extrinsic MyD88 signaling controls gene expression in cDCs and orchestrates immune responses to inhaled allergens.

Collaboration


Dive into the Keiko Nakano's collaboration.

Top Co-Authors

Avatar

Donald N. Cook

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Hideki Nakano

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gregory S. Whitehead

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Timothy P. Moran

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Seddon Y. Thomas

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rhonda H. Wilson

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kymberly M. Gowdy

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lauranell H. Burch

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Carl D. Bortner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Gordon P. Flake

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge